28.04.2013 Views

MDT (multi-disciplinary team) guidance for managing prostate cancer

MDT (multi-disciplinary team) guidance for managing prostate cancer

MDT (multi-disciplinary team) guidance for managing prostate cancer

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>MDT</strong> (Multi-<strong>disciplinary</strong> Team)<br />

Guidance <strong>for</strong><br />

Managing Prostate Cancer<br />

Produced by:<br />

• British Uro-oncology Group (BUG)<br />

• British Association of Urological Surgeons (BAUS): Section of Oncology<br />

• British Prostate Group (BPG)<br />

This <strong>guidance</strong> has been supported by educational grants from Ferring Pharmaceuticals,<br />

Date of preparation: November 2009 GB.DOC.09.11.16<br />

2nd Edition (November 2009)<br />

Novartis and sanofi-aventis. The development and content of this <strong>guidance</strong><br />

has not been influenced in any way by the supporting companies.


Abbreviations<br />

3D-CRT: three-dimensional con<strong>for</strong>mal radiotherapy<br />

ADT: androgen deprivation therapy<br />

BPH: benign prostatic hyperplasia<br />

CAB: combined androgen blockade<br />

CHHiP: Conventional or Hypofractionated High Dose IMRT <strong>for</strong> Prostate Cancer<br />

CI: confidence interval<br />

CPA: cyproterone acetate<br />

CT: computed tomography<br />

DES: diethylstilbestrol<br />

DFS: disease-free survival<br />

DRE: digital rectal examination<br />

EBRT: external beam radiation therapy<br />

EPC: Early Prostate Cancer<br />

ERSPC: European Randomised Study of Screening <strong>for</strong> Prostate Cancer<br />

FFF: freedom from failure<br />

GnRH: gonadotrophin-releasing hormone<br />

HDR: high dose rate<br />

HIFU: high-intensity focused ultrasound<br />

HR: hazard ratio<br />

HRPC: hormone-refractory <strong>prostate</strong> <strong>cancer</strong><br />

IMRT: intensity modulated radiotherapy<br />

IPSS: International Prostate Symptom Score<br />

LDR: low dose rate<br />

LHRH: luteinising hormone releasing hormone<br />

LTAD: long-term androgen deprivation<br />

<strong>MDT</strong>: <strong>multi</strong>-<strong>disciplinary</strong> <strong>team</strong><br />

MRC: Medical Research Council<br />

MRI: magnetic resonance imaging<br />

MRS: magnetic resonance spectroscopy<br />

NCCN: National Comprehensive Cancer Network<br />

NICE: National Institute <strong>for</strong> Health and Clinical Excellence<br />

ONJ: osteonecrosis of the jaw<br />

OS: overall survival<br />

PFS: progression-free survival<br />

PLCO: Prostate, Lung, Colorectal and Ovarian<br />

ProtecT: Prostate Testing <strong>for</strong> Cancer and Treatment<br />

PSA: <strong>prostate</strong>-specific antigen<br />

RCT: randomised controlled trial<br />

SRE: skeletal-related events<br />

STAD: short-term androgen deprivation<br />

TRUS: transrectal ultrasound<br />

TURP: transurethral resection of the <strong>prostate</strong><br />

2


Integrated Care and the Multi-<strong>disciplinary</strong> Team (<strong>MDT</strong>)<br />

• The concept of integrated care is becoming increasingly accepted as a way to overcome<br />

fragmentation of patient management and to provide a consistent treatment strategy across<br />

the <strong>MDT</strong>. It also creates an optimal structure that facilitates audit and peer review.<br />

• Integration within the <strong>MDT</strong> is essential <strong>for</strong> patients with <strong>prostate</strong> <strong>cancer</strong> because the<br />

collaboration between <strong>MDT</strong> members (Table 1) is central to the treatment strategy, with ongoing<br />

support from the wider <strong>team</strong> to manage pain and the adverse effects of therapy. By being familiar<br />

with the complete spectrum of management strategies, the <strong>MDT</strong> can assist patients in making<br />

treatment decisions that are specific <strong>for</strong> their individual disease state, co-morbid conditions,<br />

age and lifestyle.<br />

Table 1: The make-up of the <strong>MDT</strong> in the <strong>prostate</strong> <strong>cancer</strong> setting<br />

Urological surgeons Oncology and urology nurse specialists<br />

Clinical and medical oncologists (Palliative care specialist)<br />

<strong>MDT</strong> co-ordinator and secretarial support (Oncology pharmacists)<br />

Radiologists (Social workers)<br />

Histopathologists (Dieticians)<br />

• According to the UK-based Integrated Care Network, the move to true integrated practice can<br />

add value in the following ways: 1<br />

o Changing the identity or branding of a service to create more positive user responses and<br />

staff allegiances, enabling a clear break with the past.<br />

o Securing organisational efficiencies, <strong>for</strong> example, in the shape of shared support services,<br />

integrated management, innovative administrative processes and emerging hybrid roles.<br />

o Defining a focus <strong>for</strong> action that includes clearer processes of accountability and is less prone<br />

to distraction by wider organisational concerns.<br />

o Introducing more robust arrangements <strong>for</strong> <strong>team</strong>-working and leadership-working in<br />

challenging times.<br />

o Creating new opportunities <strong>for</strong> investment, <strong>for</strong> example, in IT systems, and opening access to<br />

new sources of funding.<br />

• The algorithms presented in this <strong>guidance</strong> provide a single framework that is adapted <strong>for</strong> each<br />

major category of <strong>prostate</strong> <strong>cancer</strong>: localised, locally advanced and advanced (Figure 1).<br />

• The treatment algorithms presented in this document (Figures 2−4) represent a management<br />

structure that goes beyond a simple co-ordinated system and will work most efficiently when<br />

the <strong>MDT</strong> is functioning as a single integrated unit.<br />

3


Figure 1: Summary of the definition of <strong>prostate</strong> <strong>cancer</strong> stages<br />

LOCALISED<br />

DISEASE<br />

T1/T2<br />

No/Mo<br />

Low Risk<br />

Gleason grade ≤6<br />

PSA ≤10<br />

Intermediate Risk<br />

Gleason grade 7<br />

and/or<br />

PSA (10-20)<br />

High Risk<br />

Gleason grade ≥8<br />

and/or<br />

PSA ≥20<br />

LOCALLY<br />

ADVANCED<br />

DISEASE<br />

(Non-metastatic)<br />

T1/T2 N+ Mo<br />

T3/T4 No/N+ Mo<br />

Failed local Rx<br />

with rising PSA<br />

and/or local<br />

recurrence+ Mo<br />

ADVANCED<br />

DISEASE<br />

(Metastatic)<br />

Proven M1<br />

4


Figure 2: Treatment algorithm <strong>for</strong> localised disease<br />

Key Questions <strong>for</strong> the <strong>MDT</strong><br />

• TNM stage?<br />

• Gleason grade?<br />

• PSA/PSA Kinetics?<br />

• Age/co-morbidity/life<br />

expectancy?<br />

• Symptoms:<br />

– Bowel?<br />

– Urine IPSS score?<br />

• Sexual function?<br />

• Family history?<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

Low Risk<br />

Gleason grade ≤6<br />

PSA ≤10<br />

Active surveillance Radical <strong>prostate</strong>ctomy<br />

Radical <strong>prostate</strong>ctomy<br />

External beam radiotherapy<br />

(EBRT) +/– neo-adjuvant<br />

and concurrent hormone<br />

therapy<br />

Low dose rate (LDR)<br />

brachytherapy +/– neoadjuvant<br />

hormone therapy<br />

Watchful waiting<br />

Novel therapies<br />

<strong>MDT</strong><br />

Diagnostic Tests<br />

• Digital rectal exam (DRE)<br />

• PSA<br />

• Transrectal ultrasound<br />

(TRUS)/biopsy<br />

• MRI/CT pelvic scan*<br />

• Bone scan*<br />

(*Not mandatory <strong>for</strong> low-risk patients)<br />

MANAGEMENT OPTIONS<br />

Intermediate Risk<br />

Gleason grade 7<br />

and/or PSA (10-20)<br />

EBRT +/– neo-adjuvant,<br />

concurrent or adjuvant<br />

hormone therapy<br />

LDR brachytherapy<br />

+/– EBRT<br />

+/– hormone therapy<br />

Watchful waiting<br />

Novel therapies<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

Key Discussion Points<br />

with the Patient<br />

• Survival/prognosis?<br />

• Treatment options?<br />

• Treatment side-effects?<br />

• Impact on quality of life?<br />

• Importance of:<br />

– Psychological impact<br />

to patient and family?<br />

– Family history?<br />

– Clinical trials?<br />

– Sexual function?<br />

– Urinary function?<br />

– Bowel function?<br />

– Physical strength, energy?<br />

– Level of activity?<br />

– Accessibility to Rx?<br />

High Risk<br />

Gleason grade ≥8<br />

and/or PSA ≥20<br />

Selected Selected<br />

For treatment<br />

approach see<br />

locally advanced<br />

management<br />

5


Figure 3: Treatment algorithm <strong>for</strong> locally advanced (non-metastatic) disease<br />

Key Questions <strong>for</strong> the <strong>MDT</strong><br />

• TNM stage?<br />

• Gleason grade?<br />

• PSA/PSA Kinetics?<br />

• Age/co-morbidity/life<br />

expectancy?<br />

• Symptoms:<br />

– Bowel?<br />

– Urine IPSS score?<br />

• Sexual function?<br />

• Family history?<br />

• Bone pain?<br />

• Osteoporosis risk?<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

High-risk localised<br />

<strong>MDT</strong><br />

Diagnostic Tests<br />

• Digital rectal exam (DRE)<br />

• PSA<br />

• Transrectal ultrasound<br />

(TRUS)/biopsy<br />

• MRI/CT pelvic scan<br />

• Bone scan<br />

• Consider lymph node sampling<br />

(if this will determine changes<br />

in management approach)<br />

MANAGEMENT OPTIONS 1<br />

T1/T2 N+ Mo<br />

T3/T4 No/N+ Mo<br />

External beam radiotherapy (EBRT)<br />

+/- HDR brachytherapy boost<br />

+/-neo-adjuvant, concomitant<br />

and adjuvant hormone therapy<br />

Hormone therapy alone<br />

Watchful waiting<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

Key Discussion Points<br />

with the Patient<br />

• Survival/prognosis?<br />

• Treatment options?<br />

• Treatment side-effects?<br />

• Impact on quality of life?<br />

• Importance of:<br />

– Psychological impact<br />

to patient and family?<br />

– Family history?<br />

– Clinical trials?<br />

– Sexual function?<br />

– Urinary function?<br />

– Bowel function?<br />

– Physical strength, energy?<br />

– Level of activity?<br />

– Accessibility to Rx?<br />

6


Figure 3: Treatment algorithm <strong>for</strong> locally advanced (non-metastatic) disease (cont.)<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

Post-radical <strong>prostate</strong>ctomy<br />

MANAGEMENT OPTIONS 2<br />

Failed local Rx with rising PSA<br />

and/or local recurrence+ Mo<br />

Active surveillance/watchful waiting Active surveillance/watchful waiting<br />

Hormone therapy alone Hormone therapy alone<br />

External beam radiotherapy<br />

(EBRT) +/– concomitant +/–<br />

adjuvant hormone therapy<br />

Post-radical radiotherapy/<br />

brachytherapy<br />

Salvage <strong>prostate</strong>ctomy<br />

Novel local salvage therapy<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

7


Figure 4: Treatment algorithm <strong>for</strong> advanced (metastatic) disease<br />

Key Questions <strong>for</strong> the <strong>MDT</strong><br />

• TNM stage?<br />

• Gleason grade?<br />

• PSA/PSA Kinetics?<br />

• Age/co-morbidity/life<br />

expectancy?<br />

• Symptoms:<br />

– Bowel?<br />

– Urine IPSS score?<br />

• Sexual function?<br />

• Family history?<br />

• Bone pain?<br />

• Osteoporosis risk?<br />

• Palliative care referral?<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

Palliative Care<br />

• Pain control<br />

• Local radiotherapy<br />

Bone pain<br />

Spinal cord compression<br />

Nerve root compression<br />

• Continual assessment<br />

Second-/third-line if<br />

androgen sensitive?<br />

<strong>MDT</strong><br />

Diagnostic Tests<br />

• Digital rectal exam (DRE)<br />

• PSA<br />

• Transrectal ultrasound (TRUS)/<br />

biopsy (non mandatory if PSA<br />

>100 and radiological evidence<br />

of metastases)<br />

• Biochemistry screen<br />

• Full blood count<br />

• Bone scan<br />

• MRI/CT pelvic scan<br />

MANAGEMENT OPTIONS<br />

Proven M1/M2<br />

First line hormone therapy<br />

Significant rise in PSA Clinical/<br />

radiological progression<br />

Observation<br />

Second-/third-line hormone<br />

therapy<br />

Further progression<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

Key Discussion Points<br />

with the Patient<br />

• Survival/prognosis?<br />

• Treatment options?<br />

• Treatment side-effects?<br />

• Impact on quality of life?<br />

• Importance of:<br />

– Psychological impact<br />

to patient and family?<br />

– Family history?<br />

– Clinical trials?<br />

– Sexual function?<br />

– Urinary function?<br />

– Bowel function?<br />

– Physical strength, energy?<br />

– Level of activity?<br />

– Accessibility to Rx?<br />

HRPC<br />

Clinical trials<br />

Chemotherapy<br />

Strontium<br />

Bisphosphonate<br />

8


Integrated care and clinical governance<br />

• The effective functioning of the <strong>MDT</strong> and tailored care pathways <strong>for</strong> patients will support the<br />

(now routine) clinical governance procedures implemented throughout the NHS. Traditionally,<br />

clinical governance relates to a single organisation or service and this can raise challenges, with<br />

the recognition that patients require management across different organisations and services.<br />

There<strong>for</strong>e, it is appropriate to apply the principles of clinical governance to individual patients or<br />

groups of patients.<br />

• The focus should be on optimum patient satisfaction and care, rather than on per<strong>for</strong>mance of<br />

the NHS institution. The <strong>MDT</strong> and development of organised pathways (allowing <strong>for</strong> individual<br />

clinical discretion and patient involvement in decision-making) ensures that the patient’s journey<br />

is monitored and assessed as a single entity.<br />

9


Approach within the <strong>MDT</strong><br />

Key questions <strong>for</strong> the <strong>MDT</strong><br />

• TNM stage?<br />

• Gleason grade?<br />

• Prostate-specific antigen (PSA) concentration?<br />

• Risk group (according to National Comprehensive Cancer Network [NCCN] guidelines)?<br />

• Age/co-morbidity/life expectancy?<br />

• Symptoms:<br />

o International Prostate Symptom Score (IPSS), bowel, urinary?<br />

• Sexual function?<br />

• Bone pain?<br />

• Osteoporosis risk?<br />

• Family history?<br />

• Clinical trials?<br />

• The <strong>MDT</strong> is an essential part of <strong>cancer</strong> management. However, there are often difficulties in<br />

knowing which patients to discuss, ensuring that their details and diagnoses are available,<br />

and keeping a record of decisions made at the meetings.<br />

• <strong>MDT</strong>s have repeatedly been endorsed as the principal mechanism <strong>for</strong> ensuring that all relevant<br />

disciplines and professional groups contribute to, and participate in, decisions regarding the<br />

clinical management of patients. 2<br />

• <strong>MDT</strong>-working is positively related to a range of measures of effectiveness, including the quality<br />

of clinical care.<br />

• It is important to emphasise the distinction between management and administration.<br />

• A central concept of integrated care is to rein<strong>for</strong>ce the role of the <strong>MDT</strong> (working as a single unit),<br />

but with enough clinical freedom to tailor management strategies to the needs of individual<br />

patients.<br />

• Treatment strategies are influenced by the stage of disease and by an interaction between<br />

the risk of disease progression, survival and key patient characteristics, such as age, lifestyle<br />

and general health. The discussion of these factors is of crucial importance in determining the<br />

most appropriate way <strong>for</strong>ward. For example, age and the presence of co-morbidities may be a<br />

restrictive factor when considering surgery.<br />

• The case notes, pathology reports, test results and radiology <strong>for</strong> each patient must be available<br />

to be discussed at the meeting. The <strong>MDT</strong> must also ensure that the patient has the fullest possible<br />

role in determining treatment − the importance of this cannot be overstated. Patient preference<br />

should be discussed within the <strong>MDT</strong>. Although the majority of men with <strong>prostate</strong> <strong>cancer</strong> want to<br />

be involved in treatment decisions, an estimated one in five of all patients does not raise, or really<br />

understand, the potential issues and associated side-effects of treatments and alternatives that<br />

may be available to them. 3<br />

• The possibility of including a patient in a relevant clinical trial should not be <strong>for</strong>gotten.<br />

10


Approach to the Patient<br />

Key points <strong>for</strong> discussion with the patient<br />

• Survival prognosis?<br />

• Treatment options?<br />

• Treatment side-effects?<br />

• Impact on quality of life?<br />

• Importance of:<br />

o Sexual function?<br />

o Urinary function?<br />

o Bowel function?<br />

o Physical strength, energy?<br />

o Level of activity?<br />

o Accessibility to prescribed drugs?<br />

o Psychosocial impact on them and their family?<br />

• Family history?<br />

• Clinical trials?<br />

The patient’s expectations<br />

The patient should have the right to discuss their treatment with<br />

appropriately trained members of the <strong>MDT</strong><br />

• After a diagnosis of <strong>prostate</strong> <strong>cancer</strong>, most men will want to have some involvement in the<br />

decisions concerning their care. The following aspects have been found to be important: 4<br />

o Honesty about the severity of the <strong>cancer</strong> and their prognosis<br />

o Discussion of the best treatment options<br />

o The clinician being up-to-date on ongoing and recent research<br />

o Disclosing all treatment options<br />

o How <strong>cancer</strong> may affect their daily functioning<br />

• It is essential that the patient and healthcare professionals discuss the likelihood of adverse<br />

events associated with each treatment option and implications <strong>for</strong> their future lifestyle when<br />

determining management strategies.<br />

• The patient and his partner, family and/or other carers should be fully in<strong>for</strong>med about care and<br />

treatment options and there<strong>for</strong>e able to make appropriate decisions based upon the choices<br />

offered by their healthcare professionals. For example, the choice between radical treatment and<br />

active surveillance may be influenced by a patient’s desire to retain sexual activity, physical energy<br />

and quality of life.<br />

• Patients should be in<strong>for</strong>med and advised regarding the available treatment options and the<br />

potential effects of these on their lifestyle and quality of life.<br />

11


Discussing evidence with patients<br />

There is a lack of evidence to guide how healthcare professionals can most effectively share clinical<br />

data with those patients facing treatment decisions. However, basing recommendations largely on<br />

relevant clinical studies and expert opinion, it is possible to achieve five communication objectives<br />

when framing and communicating clinical evidence.<br />

1. Understand the patient’s experience, expectations and preferences<br />

2. Build partnerships with the patient and carer<br />

3. Provide evidence and discuss uncertainties and side-effects<br />

4. Present recommendations<br />

5. Check <strong>for</strong> understanding and agreement<br />

12


Assessment and Diagnosis<br />

Screening<br />

• PSA screening remains a relatively contentious subject in the field of <strong>prostate</strong> <strong>cancer</strong>. It is felt that<br />

robust evidence is lacking concerning whether screening results in a reduction in mortality from<br />

the disease.<br />

• Three ongoing large, randomised, controlled clinical trials are evaluating the value of PSA<br />

screening <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong>: the European Randomised Study of Screening <strong>for</strong> Prostate Cancer<br />

(ERSPC), 5 the Prostate, Lung, Colorectal and Ovarian (PLCO) <strong>cancer</strong> screening trial in the US6<br />

and the UK-based Prostate Testing <strong>for</strong> Cancer and Treatment (ProtecT) study. 7 The first reports<br />

from these trials have been published and have added further in<strong>for</strong>mation to the PSA screening<br />

debate:<br />

o The PLCO study reported no mortality benefit with the combination of PSA screening and<br />

digital rectal examination (DRE) during a median follow-up of 7 years. 6<br />

o In contrast, the ERSPC trial found that PSA screening was associated with a 20% relative<br />

reduction in <strong>prostate</strong> <strong>cancer</strong> mortality at a median follow-up of 9 years, equivalent to the<br />

prevention of approximately 7 <strong>prostate</strong> <strong>cancer</strong> deaths per 10,000 men screened. This mortality<br />

benefit was associated with a high risk of overdiagnosis, with nearly 76% of men who<br />

underwent a biopsy following an elevated PSA value having a false positive result. 5<br />

o ProtecT has demonstrated a benefit of repeat PSA testing in reducing the risk of high-grade<br />

<strong>prostate</strong> <strong>cancer</strong> in men with an initial PSA concentration of 3−20 ng/ml. 7<br />

• Quality of life and cost-effectiveness analyses from the ERSPC and PLCO trials, along with<br />

mortality results from ProtecT are needed to help resolve the ongoing PSA screening debate.<br />

Risk factors <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong><br />

The risk factors <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong> are generally well-documented, but are highlighted here <strong>for</strong><br />

completeness of the Guidance.<br />

• Age<br />

o Relatively rare in men under the age of 50 years.<br />

o Incidence increases in those over 60 years.<br />

• Race<br />

o A higher incidence of the disease is seen in African-Caribbean, African-American and West<br />

African races. The UK PROCESS study is compiling equivalent UK data. 8<br />

o Men of Chinese and Japanese origin have a low incidence of disease.<br />

• Geography<br />

o The highest incidence of <strong>prostate</strong> <strong>cancer</strong> is currently seen in North America and<br />

Northern Europe.<br />

• Family history<br />

o Men with a first-degree relative affected by <strong>prostate</strong> <strong>cancer</strong> have a relative risk of<br />

developing the disease themselves 2-fold greater than men with no relatives affected. 9<br />

o Those men with an affected second-degree relative have an increased relative risk<br />

of 1.7 of developing the disease.<br />

o Men with both a first- and second-degree relative affected have an increased relative<br />

risk of 8.8 of developing the disease.<br />

13


o There is also some evidence to show a link between an increased risk of <strong>prostate</strong> <strong>cancer</strong><br />

where there is a family history of breast, ovarian, bladder or kidney <strong>cancer</strong>. 10<br />

o The UK Familial Prostate Cancer Study is currently looking at the genetics of the disease<br />

with possible sites of interest lying on chromosomes 2, 5, Y and loss of heterozygosity at<br />

10q and 16q.<br />

Diagnostic tests<br />

DRE<br />

• The DRE remains valid as an initial method <strong>for</strong> assessing the <strong>prostate</strong>; however, DRE findings<br />

should not be regarded as a fail-safe test.<br />

PSA<br />

• As an independent variable, PSA concentrations are a better predictor of <strong>cancer</strong> than suspicious<br />

11, 12<br />

findings on DRE or transrectal ultrasound (TRUS).<br />

• Serum concentrations of PSA can be elevated in the presence of benign prostatic hyperplasia<br />

(BPH), prostatitis and other non-malignant conditions. Furthermore, there is, as yet, no<br />

recommendation <strong>for</strong> the optimal PSA threshold value that most effectively avoids the detection<br />

13, 14<br />

of insignificant <strong>cancer</strong>s that are unlikely to be life-threatening.<br />

• While PSA concentrations generally increase with advancing disease stage, the ability of PSA levels<br />

to accurately predict pathological stage in any one individual is low. 15−17<br />

• Asymptomatic patients who request a PSA test should be counselled be<strong>for</strong>e the procedure<br />

<strong>for</strong> the following reasons: 18<br />

o Although the test may detect a <strong>cancer</strong> at a stage where curative treatment can be offered,<br />

PSA will fail to detect some early tumours.<br />

o A PSA test may detect early <strong>prostate</strong> <strong>cancer</strong> in an estimated 5% of men aged 50−65 years.<br />

o Treatment of early <strong>prostate</strong> <strong>cancer</strong> can put the patient at some risk and may not necessarily<br />

improve life expectancy.<br />

Factors affecting PSA concentrations are summarised below.<br />

Age and race<br />

Table 2: Age-specific PSA (ng/ml) reference ranges, by race 19<br />

Age (years) White Black Latino Asian<br />

40−49 0−2.3 0−2.7 0−2.1 0−2.0<br />

50−59 0−3.8 0−4.4 0−4.3 0−4.5<br />

60−69 0−5.6 0−6.7 0−6.0 0−5.5<br />

70−79 0−6.9 0−7.7 0−6.6 0−6.8<br />

Biopsy/transurethral resection of the <strong>prostate</strong> (TURP) can cause an increase in PSA <strong>for</strong> a variable time<br />

period (4−12 weeks). 20<br />

Prostatitis can cause an increase in PSA concentration, which can be reduced to within a normal range<br />

21, 22<br />

with antibiotic treatment.<br />

14


Prostate size – a benignly enlarged gland can influence PSA concentrations.<br />

Infection – elevated PSA levels can be seen beyond 6 months in up to 50% of patients when associated<br />

with febrile urinary tract infections.<br />

Free and complexed PSA should be understood. Catalona et al. conclude that percentage free PSA<br />

is most useful in men with a PSA concentration in the range 2−15 ng/ml (Table 3); the higher the<br />

percentage of free PSA the lower the probability of <strong>cancer</strong>. 23<br />

Table 3: Probability of <strong>prostate</strong> <strong>cancer</strong> based on total and percentage free PSA 23<br />

Total PSA (ng/ml)<br />

PSA density i.e. PSA level (ng/ml)<br />

TRUS-determined <strong>prostate</strong> volume (ml)<br />

Probability of <strong>cancer</strong> (%)<br />

0−2 ~1<br />

2−4 15<br />

4−10 25<br />

>10 >50<br />

Free PSA (%)<br />

0−10 56<br />

10−15 28<br />

15−20 20<br />

20−25 16<br />

>25 8<br />

May be helpful in differentiating BPH from <strong>prostate</strong> <strong>cancer</strong> in patients who have a normal DRE with<br />

a PSA 4−10ng/ml. A PSA density >0.15 may suggest <strong>prostate</strong> <strong>cancer</strong>.<br />

PSA velocity can be valuable in the follow-up of men with a normal PSA but prior negative biopsies.<br />

Velocity is measured by a change in PSA concentration in three consecutive measurements taken at<br />

6-monthly intervals. A change in PSA concentration of >0.75 ng/ml per year is more likely to indicate<br />

<strong>prostate</strong> <strong>cancer</strong> than BPH. The usefulness of PSA velocity in those with a PSA concentration >10 ng/ml<br />

is unknown. 24<br />

TRUS<br />

• TRUS detects 50% more patients with <strong>prostate</strong> <strong>cancer</strong> than physical examination alone, 25, 26 but<br />

the ultrasonic appearance of <strong>prostate</strong> <strong>cancer</strong> is variable and only a very small number of <strong>cancer</strong>s<br />

are detected if a DRE and PSA test are normal. 26−28 There<strong>for</strong>e, TRUS is mainly used to aid biopsy.<br />

15


Biopsy and tumour grading<br />

• The National Institute <strong>for</strong> Health and Clinical Excellence (NICE) <strong>prostate</strong> <strong>cancer</strong> guideline<br />

recommends that the serum PSA level alone should not automatically lead to a <strong>prostate</strong> biopsy. 29<br />

It states that to help men decide whether to have a <strong>prostate</strong> biopsy, healthcare professionals<br />

should discuss with them their PSA level, DRE findings (including an estimate of <strong>prostate</strong> size)<br />

and co-morbidities, together with their risk factors (including increasing age and black African<br />

and black Caribbean ethnicity) and any history of a previous negative <strong>prostate</strong> biopsy.<br />

• NICE further highlights that men and their partners or carers should be given in<strong>for</strong>mation,<br />

support and adequate time to decide whether or not they wish to undergo <strong>prostate</strong> biopsy. 29<br />

Men will need to comprehend the potential risks (such as potentially living with a diagnosis of<br />

<strong>prostate</strong> <strong>cancer</strong> that is deemed clinically insignificant) and the benefits of <strong>prostate</strong> biopsy.<br />

• Where biopsy is indicated, a minimum of 10 biopsies should be obtained, according to the volume<br />

of the <strong>prostate</strong>. Biopsies should be per<strong>for</strong>med under local anaesthetic and antibiotic cover. 30<br />

• A European study has reported that a <strong>prostate</strong> <strong>cancer</strong> detection rate <strong>for</strong> the first set of biopsies<br />

is 24% and <strong>for</strong> the second set of biopsies after a negative initial set as 13%. 31<br />

Magnetic Resonance Imaging (MRI)<br />

• The classification used <strong>for</strong> staging <strong>prostate</strong> <strong>cancer</strong> is the internationally approved system<br />

recommended by the Union International Contra Cancer (available at: www.uicc.org).<br />

• TNM staging, Gleason score, and PSA concentration facilitate estimation of the risk of<br />

extracapsular disease and lymph node metastases. Pelvic staging is required <strong>for</strong> those of high<br />

or intermediate risk (according to NCCN classification). MRI is the preferred option to stage pelvic<br />

lesions and where MRI is contraindicated, computed tomography (CT) should be used. 29<br />

• Body coil MRI is sensitive and specific in identifying extracapsular extension of <strong>prostate</strong> <strong>cancer</strong><br />

in patients with high- or intermediate-risk disease. 32<br />

• NICE concludes: 29<br />

o MRI is now the most accurate and commonly-used imaging technique <strong>for</strong> tumour-staging<br />

men with <strong>prostate</strong> <strong>cancer</strong>. Many of the original publications used now outdated MRI<br />

technology, and the accuracy reported <strong>for</strong> MRI is improving, typically with endorectal coil<br />

imaging at 1.5 Tesla.<br />

o After transrectal <strong>prostate</strong> biopsy, intra-prostatic haematoma can affect image interpretation<br />

<strong>for</strong> at least 4 weeks.<br />

o Magnetic resonance spectroscopy (MRS) is an experimental technique based on the<br />

concentration of metabolites such as choline and citrate in the <strong>prostate</strong> gland. Prostate <strong>cancer</strong><br />

alters the concentrations of these metabolites and this may be used to find areas of tumour<br />

activity. MRS is not recommended <strong>for</strong> men with <strong>prostate</strong> <strong>cancer</strong> except in the context of a<br />

clinical trial.<br />

Bone scans and lymph node sampling<br />

• Bone scans (particularly in patients with PSA concentration >20 ng/ml) and lymph node biopsies<br />

are also important in the assessment process. A PSA concentration of


Key Questions <strong>for</strong> the <strong>MDT</strong><br />

• TNM stage?<br />

• Gleason grade?<br />

• PSA/PSA Kinetics?<br />

• Age/co-morbidity/life<br />

expectancy?<br />

• Symptoms:<br />

– Bowel?<br />

– Urine IPSS score?<br />

• Sexual function?<br />

• Family history?<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

Localised Disease: Management Options<br />

Figure 2: Treatment algorithm <strong>for</strong> localised disease<br />

Low Risk<br />

Gleason grade ≤6<br />

PSA ≤10<br />

Active surveillance Radical <strong>prostate</strong>ctomy<br />

Radical <strong>prostate</strong>ctomy<br />

External beam radiotherapy<br />

(EBRT) +/– neo-adjuvant<br />

and concurrent hormone<br />

therapy<br />

Low dose rate (LDR)<br />

brachytherapy +/– neoadjuvant<br />

hormone therapy<br />

Watchful waiting<br />

Novel therapies<br />

<strong>MDT</strong><br />

Diagnostic Tests<br />

• Digital rectal exam (DRE)<br />

• PSA<br />

• Transrectal ultrasound<br />

(TRUS)/biopsy<br />

• MRI/CT pelvic scan*<br />

• Bone scan*<br />

(*Not mandatory <strong>for</strong> low-risk patients)<br />

MANAGEMENT OPTIONS<br />

Intermediate Risk<br />

Gleason grade 7<br />

and/or PSA (10-20)<br />

EBRT +/– neo-adjuvant,<br />

concurrent or adjuvant<br />

hormone therapy<br />

LDR brachytherapy<br />

+/– EBRT<br />

+/– hormone therapy<br />

Watchful waiting<br />

Novel therapies<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

Key Discussion Points<br />

with the Patient<br />

• Survival/prognosis?<br />

• Treatment options?<br />

• Treatment side-effects?<br />

• Impact on quality of life?<br />

• Importance of:<br />

– Psychological impact<br />

to patient and family?<br />

– Family history?<br />

– Clinical trials?<br />

– Sexual function?<br />

– Urinary function?<br />

– Bowel function?<br />

– Physical strength, energy?<br />

– Level of activity?<br />

– Accessibility to Rx?<br />

High Risk<br />

Gleason grade ≥8<br />

and/or PSA ≥20<br />

Selected Selected<br />

For treatment<br />

approach see<br />

locally advanced<br />

management<br />

17


The following <strong>guidance</strong> <strong>for</strong> <strong>managing</strong> localised <strong>prostate</strong> <strong>cancer</strong> focuses on low- and intermediate-risk<br />

categories, defined here as: 34<br />

• Low risk (T1c/T2a; Gleason grade ≤6; PSA concentration ≤10 ng/ml)<br />

• Intermediate risk (T2b; Gleason grade 7 and/or PSA concentration: >10 and ≤20 ng/ml)<br />

In the proposed management algorithms, high-risk localised disease falls more naturally into<br />

management of locally advanced disease.<br />

Patient choice and the presence or absence of co-morbidities should be an essential component<br />

of management decisions in men with localised disease. Decisions concerning the choice of radical<br />

treatments need to be carefully balanced with the different options available and the impact of such<br />

treatments on a patient’s co-morbidities.<br />

In this section available evidence <strong>for</strong> the following management approaches is outlined:<br />

• Active surveillance<br />

• Watchful waiting<br />

• Radical <strong>prostate</strong>ctomy<br />

• External Beam Radiation Therapy (EBRT)<br />

• Low dose rate brachytherapy<br />

• Neoadjuvant/adjuvant hormone therapy<br />

• Novel therapies<br />

There is no consistent evidence <strong>for</strong> superiority of any of the three main radical treatments: radical<br />

<strong>prostate</strong>ctomy, EBRT or brachytherapy.<br />

18


Active surveillance<br />

Overview<br />

• Active surveillance is an approach to the management of early <strong>prostate</strong> <strong>cancer</strong> in which the choice<br />

between curative treatment and observation is based on evidence of disease progression (PSA<br />

kinetics or repeat biopsy findings) during a period of close monitoring. The aim is to reduce the<br />

burden of treatment side-effects without compromising survival.<br />

• Patients suitable <strong>for</strong> active surveillance are those with low-risk localised disease who are fit <strong>for</strong><br />

radical treatment. Ongoing prospective studies of active surveillance have shown that 60−80%<br />

of such men will avoid the need <strong>for</strong> treatment, and that 100% <strong>prostate</strong> <strong>cancer</strong>-specific survival<br />

35, 36<br />

at 10 years is achievable.<br />

• Active surveillance should be distinguished from watchful waiting. Traditional watchful waiting<br />

involves relatively lax observation with late, palliative treatment <strong>for</strong> those who develop symptoms<br />

of progressive disease. In contrast, active surveillance involves close monitoring with early, radical<br />

treatment in those with signs of disease progression.<br />

Patient selection<br />

• Low-risk, clinically localised <strong>prostate</strong> <strong>cancer</strong><br />

o Clinical stage T1c/2a<br />

o Gleason grade ≤3+4<br />

o PSA concentration


• Van As et al. reported the outcome of 326 men recruited to the Royal Marsden active surveillance<br />

study from 2002 to 2006, at a median follow-up of 22 months. 35 Median age was 67 years,<br />

and median initial PSA concentration 6.4 ng/ml. Sixty-five patients (20%) had deferred radical<br />

treatment, 16 (5%) changed to watchful waiting because of increasing co-morbidity, 7 (2%) died<br />

of other causes, and 238 (73%) remain on surveillance. There were no deaths from <strong>prostate</strong> <strong>cancer</strong><br />

and no men had developed metastatic disease.<br />

• It is possible that targeted radical treatment, based on an active surveillance strategy, will be as<br />

effective, and considerably less morbid, than radical treatment <strong>for</strong> all cases. The ProSTART phase III<br />

trial will compare the long term outcomes of active surveillance with those of immediate radical<br />

treatment, and aims to recruit approximately 2100 patients.<br />

20


Watchful waiting<br />

Overview<br />

• Watchful waiting is an approach to the management of localised <strong>prostate</strong> <strong>cancer</strong> that aims to<br />

avoid treatment, or delay it <strong>for</strong> as long as possible. Patients who eventually develop symptoms<br />

of progressive <strong>prostate</strong> <strong>cancer</strong> receive palliative treatment, usually with hormone therapy.<br />

• Watchful waiting is particularly suitable <strong>for</strong> patients aged over 75 years or younger men with<br />

significant co-morbidities.<br />

• Watchful waiting should be distinguished from active surveillance. Conventional watchful waiting<br />

involves relatively lax observation with late, palliative treatment <strong>for</strong> those who develop symptoms<br />

of progressive disease. In contrast, active surveillance involves close monitoring with early, radical<br />

treatment in those with signs of progression.<br />

Patient selection<br />

• Asymptomatic clinically localised <strong>prostate</strong> <strong>cancer</strong><br />

o Clinical stage T1−3<br />

o Gleason score ≤7<br />

o Any PSA concentration<br />

o Not suitable <strong>for</strong> radical treatment (usually by virtue of older age or co-morbidities)<br />

Side-effects<br />

• Uncertainty<br />

Clinical evidence<br />

• The NICE clinical guideline confirms a lack of evidence <strong>for</strong> watchful waiting and the Guideline<br />

Development Group reached a consensus that the recommendation from NICE would avoid<br />

unnecessary investigations: 29<br />

o Men with localised <strong>prostate</strong> <strong>cancer</strong> who have chosen a watchful waiting regimen and who<br />

have evidence of significant disease progression (that is, rapidly rising PSA level or bone pain)<br />

should be reviewed by a member of the urological <strong>cancer</strong> <strong>MDT</strong>.<br />

21


Radical treatments<br />

Radical <strong>prostate</strong>ctomy<br />

There are now four approaches to per<strong>for</strong>ming a radical <strong>prostate</strong>ctomy: retropubic, perineal, laparoscopic<br />

and robotic. The procedure involves removal of the whole <strong>prostate</strong> and the seminal vesicles. The most<br />

commonly used approaches are retropubic and perineal, but laparoscopic and robotic techniques are<br />

becoming more frequently adopted. The newer approaches have the advantage of reduced blood loss<br />

and shorter inpatient stays.<br />

Overview<br />

• In 1997, Selley et al. reviewed a total of 17 studies (two randomised controlled trials [RCTs] and<br />

15 observational studies involving a total of 5410 patients) to investigate the efficacy of radical<br />

<strong>prostate</strong>ctomy <strong>for</strong> men with localised <strong>prostate</strong> <strong>cancer</strong>. Cancer-specific survival after 10 years of followup<br />

ranged from 86% to 91%, with clinical disease-free survival (DFS) ranging from 57% to 83%. 38<br />

Patient selection<br />

• Anaesthetic fitness<br />

• At least 10 years’ life expectancy<br />

• Complications increase and benefits decrease in patients aged >70 years<br />

Side-effects of treatment<br />

• Based on the systematic review by Selley et al., the following side-effects should be considered: 38<br />

o Operative and post-operative mortality: 0.2−1.2%<br />

o Sexual dysfunction: 51−61%<br />

o Incontinence (mild stress): 4−21%<br />

o Incontinence (total): 0−7%<br />

Clinical evidence<br />

• One randomised trial has compared radical <strong>prostate</strong>ctomy with watchful waiting in localised<br />

<strong>prostate</strong> <strong>cancer</strong>. 39<br />

o The trial randomised 695 men with localised disease between radical <strong>prostate</strong>ctomy and<br />

watchful waiting.<br />

o At a median follow-up of 8.2 years, randomisation to radical <strong>prostate</strong>ctomy was associated with<br />

a benefit both in terms of disease-specific mortality (hazard ratio [HR] 0.56; 95% confidence<br />

interval [CI]: 0.36−0.88; p=0.01) and overall mortality (HR 0.74; 95%CI: 0.56−0.99; p=0.04).<br />

o This translated into 10-year OS of 73% versus 68% (p=0.04) <strong>for</strong> radical <strong>prostate</strong>ctomy versus<br />

watchful waiting.<br />

o In terms of 10-year freedom from distant metastases, the absolute benefit of surgery versus<br />

watchful waiting was 10% (84.8% versus 74.6%; p=0.004).<br />

o However, patients need to weigh these benefits against the risk of adverse consequences of<br />

treatment. The 5% absolute improvement in 10-year survival was achieved at the expense of a<br />

35% absolute increase in the risk of erectile dysfunction and a 28% absolute increase in the risk<br />

of urinary leakage.<br />

22


o Faced with these figures, some patients would choose surgery, but many would choose<br />

conservative management. 40<br />

• The above findings are now updated to incorporate a further 3 years of follow up. At 12 years: 41<br />

o 12.5% of the surgery group and 17.9% of the watchful waiting group had died of <strong>prostate</strong><br />

<strong>cancer</strong> (difference = 5.4%, 95%CI: 0.2−11.1).<br />

o 19.3% of men in the surgery group and 26% of men in the watchful waiting group had been<br />

diagnosed with distant metastases (difference = 6.7%, 95%CI: 0.2−13.2).<br />

o This follow-up analysis was consistent with the previous findings. Radical <strong>prostate</strong>ctomy can<br />

reduce <strong>prostate</strong> <strong>cancer</strong> mortality and risk of metastases but no further benefits were seen<br />

beyond 10 years of follow up in this study.<br />

Neoadjuvant and adjuvant hormone therapy with radical <strong>prostate</strong>ctomy<br />

• At present, evidence suggests that the down-staging achieved with neoadjuvant hormone<br />

therapy does not translate into improved DFS, and there<strong>for</strong>e cannot be recommended outside<br />

of clinical trials. 42−45<br />

• Similarly, there is currently no evidence that adjuvant hormone therapy provides a survival<br />

46, 47<br />

advantage <strong>for</strong> patients with pathologically proven localised disease.<br />

Adjuvant radiotherapy after radical <strong>prostate</strong>ctomy<br />

• The EORTC 22911 study was designed to investigate benefit <strong>for</strong> immediate postoperative<br />

radiotherapy in 502 patients with pT3 disease or positive surgical margins as opposed to<br />

radiotherapy offered <strong>for</strong> biochemical or clinical relapse. 48<br />

o After a median follow up of 5 years, biochemical progression-free survival (PFS) was<br />

significantly improved in the immediate radiotherapy group (74.0% versus 52.6%;<br />

p


External Beam Radiotherapy (EBRT)<br />

Overview<br />

• Selley et al. reviewed 21 observational studies and one RCT involving radiotherapy and found that<br />

survival and recurrence rates are associated with grade and stage of the disease. The 5-year DFS<br />

<strong>for</strong> those with T1–T2 stage disease averaged 70−80%. Local progression was observed in 10−20%<br />

of these patients, while distant metastases were observed in 20−40%. 38<br />

• Nilsson et al. per<strong>for</strong>med a systematic overview of radiotherapy in <strong>prostate</strong> <strong>cancer</strong>. Data from 26<br />

non-randomised trials of conventional EBRT showed a 10-year DFS of 100%, 69% and 57% <strong>for</strong><br />

T1a, T1b and T2 stage disease, respectively. 51<br />

• Long-term follow-up after EBRT continues to demonstrate an improvement in cause-specific<br />

survival. Improved selection and technical developments in radiotherapy leading to increased<br />

doses have shown better results.<br />

Three-dimensional con<strong>for</strong>mal radiotherapy (3D-CRT)<br />

• There is evidence that increased radiation dose is associated with increased <strong>cancer</strong> cell kill <strong>for</strong> men<br />

with localised <strong>prostate</strong> <strong>cancer</strong>. However, the traditional two-dimensional technique of treatment<br />

planning and delivery is limited by the normal tissue toxicity of the surrounding structures<br />

(bladder, rectum and bowel), such that the dose that can be safely delivered to the <strong>prostate</strong><br />

by EBRT is 65−70 Gy. Several technological advances over the last 20 years have enhanced the<br />

precision of EBRT, and have resulted in improved outcomes.<br />

• The three-dimensional con<strong>for</strong>mal radiotherapy (3D-CRT) approach reduces the dose-limiting late<br />

side-effect of proctitis 52 and has allowed <strong>for</strong> dose escalation to the whole <strong>prostate</strong> to 78 Gy.<br />

Intensity Modulated Radiotherapy (IMRT)<br />

• IMRT is an advanced technique of 3D-CRT. IMRT can modify the shape and intensity of the<br />

<strong>multi</strong>ple radiotherapy beams. It is very precise in targeting the treatment area, sparing<br />

surrounding tissue. IMRT is currently under evaluation, particularly <strong>for</strong> the irradiation of pelvic<br />

lymph nodes.<br />

• IMRT may result in reduced rectal toxicity when using doses greater than 80 Gy.<br />

Dose escalation<br />

• Evidence of the benefits of dose escalation has been demonstrated <strong>for</strong> T1−T3 <strong>prostate</strong> <strong>cancer</strong> by<br />

Pollack et al. in a phase III randomised study undertaken at the MD Anderson Hospital. 53<br />

o A total of 305 men were randomised between 1993 and 1998 to compare the efficacy of 70<br />

Gy versus 78 Gy with a median follow-up of 60 months. The primary endpoint was freedom<br />

from failure (FFF), including biochemical failure, which was defined as three rises in PSA level.<br />

o The FFF rates <strong>for</strong> the 70 Gy and 78 Gy arms at 6 years were 64% and 70%, respectively (p=0.03).<br />

Dose escalation to 78 Gy preferentially benefited those with a pre-treatment PSA concentration<br />

>10 ng/ml; the FFF rate was 62% <strong>for</strong> the 78 Gy arm versus 43% <strong>for</strong> those who received 70 Gy<br />

(p=0.01). For patients with a pre-treatment PSA concentration ≤10 ng/ml, no significant doseresponse<br />

relationship was found, with an average 6-year FFF rate of about 75%.<br />

o Although no difference in OS occurred, the freedom from distant metastasis rate was<br />

higher <strong>for</strong> those with PSA levels >10 ng/ml who were treated to 78 Gy (98% versus 88%<br />

at 6 years, p=0.056).<br />

24


• Dearnaley and colleagues have reported their findings from the MRC RT01 study. 54<br />

o In this 3D-CRT trial, 843 men were randomised to a standard dose of 64 Gy compared with<br />

an escalated dose of 74 Gy, with all men also receiving neoadjuvant hormone therapy.<br />

o Patients receiving the conventional dose had 5-year biochemical PFS rates of 60% compared<br />

to 71% in the dose-escalated arm. Advantages were also seen in terms of clinical PFS and the<br />

decreased use of androgen suppression.<br />

o Although these and other studies have shown benefits from dose escalation with 3D-CRT<br />

techniques, this has been offset to a degree by a reported increase in late rectal toxicity.<br />

• Prospective non-randomised studies conducted at the Memorial Sloan Kettering <strong>cancer</strong> centre<br />

have compared the outcomes of 1100 men who received doses in the range of 64−70 Gy and<br />

76−86 Gy using IMRT. 55<br />

o The results were evaluated within prognostic risk groups (using clinical stage, Gleason grade<br />

and presenting PSA concentration). They demonstrated that increasing the dose delivered<br />

beyond 70.2 Gy in men with intermediate- and high-risk disease improved the 5-year actuarial<br />

PSA relapse-free survival rate from 50% to 70% and 21% to 47%, respectively, in these two<br />

risk categories.<br />

• IMRT has the potential to reduce late rectal toxicity as shown in a further study that reports 3-year<br />

actuarial ≥grade 2 gastrointestinal toxicity at 4%. 56<br />

• A further development under investigation involves a change in the traditional fractionation<br />

schedules. Hypofractionation may improve <strong>cancer</strong> control <strong>for</strong> the same level of radiation-related<br />

toxicity and be a more effective treatment <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong> with a predicted low alpha/<br />

beta ratio. Phase II dose escalation studies using shortened schedules of hypofractionated IMRT<br />

regimens have indicated acceptable early toxicity. 57<br />

• The CHHiP (Conventional or Hypofractionated High Dose IMRT <strong>for</strong> Prostate Cancer) study is<br />

currently recruiting patients in the UK to compare standard fractionation IMRT (74 Gy in 37<br />

fractions) to two hypofractionated IMRT regimens (60 Gy in 20 fractions or 57 Gy in 19 fractions)<br />

in combination with neoadjuvant hormone therapy. 58<br />

Patient selection<br />

• EBRT can be unsuitable <strong>for</strong> patients with bilateral hip replacement, previous radiotherapy, severe<br />

proctitis or bowel morbidity.<br />

Side-effects<br />

• Acute complications include cystitis, faecal frequency and urgency, proctitis and rectal bleeding.<br />

• Late complications occurring 3 months or later after treatment include impotence, bleeding,<br />

proctitis and diarrhoea.<br />

EBRT plus neoadjuvant hormone therapy<br />

• Neoadjuvant hormone therapy reduces <strong>prostate</strong> volume by 30−40%. 59, 60 This can reduce the size of<br />

the treatment field and as a result the level of toxicity experienced.<br />

• There are also reports of an additive or synergistic effect on tumour cell kill with combined<br />

therapy. Theories as to the mechanism of this include improved oxygenation by reducing tumour<br />

bulk and movement of hormone-responsive cells into a resting phase, which could reduce<br />

repopulation rate and enhance tumour cell death (increased apoptosis). 61<br />

• The RTOG 86-10 trial randomised 471 men with T2−T4 <strong>prostate</strong> <strong>cancer</strong> to radiotherapy +/− 4<br />

months of androgen deprivation therapy (ADT) be<strong>for</strong>e and during EBRT or to radiotherapy alone. 62<br />

25


o At median follow-up of 8.7 years, there was a trend to improved survival (8-year survival<br />

53% versus 44%, p=0.1) <strong>for</strong> those treated by hormone therapy with radiotherapy, which was<br />

significant <strong>for</strong> the subgroup with Gleason grade 2−6 disease (70% versus 52%, p=0.015). 62<br />

o Ten-year OS estimates (43% versus 34%) and median survival times (8.7 versus 7.3 years)<br />

favoured combined therapy with hormones and radiation compared to radiation treatment<br />

alone; however, these differences did not reach statistical significance (p=0.12).<br />

o There was a statistically significant improvement in 10-year disease-specific mortality<br />

(23% versus 36%; p=0.01), distant metastases (35% versus 47%; p=0.006), DFS (11% versus<br />

3%; p


Low dose rate (LDR) brachytherapy<br />

Overview<br />

• In 2005, NICE reviewed the medical literature on LDR brachytherapy and concluded that, in the<br />

absence of randomised trials, the results of LDR brachytherapy are comparable to those achieved<br />

with surgery or EBRT in well-selected patients. 66<br />

• Suitable patients include those with localised disease (up to T2a) with a Gleason grade ≤6, and a<br />

PSA concentration ≤10 ng/ml. Patients with significant urinary symptoms or post-TURP may not be<br />

suitable.<br />

• Brachytherapy is as effective as radical <strong>prostate</strong>ctomy in patients with low-risk localised disease. 67<br />

• In intermediate-risk localised disease, the comparison is less clear, because many studies have<br />

added EBRT in combination. 68<br />

• Brachytherapy is a single-step procedure following a spinal or general anaesthetic.<br />

Brachytherapy plus EBRT<br />

• In a matched-pair analysis, the 5-year biochemical failure-free survival rate was 86% <strong>for</strong> patients<br />

treated with EBRT and LDR brachytherapy, and 72% <strong>for</strong> patients treated with EBRT alone (p=0.03).<br />

Both treatments were associated with comparable incidences of late genitourinary side-effects<br />

(18−19%). Late rectal toxicity decreased by 15% in patients treated with EBRT and brachytherapy<br />

(p=0.0003). 69<br />

Brachytherapy plus neoadjuvant hormone therapy<br />

• The role of neoadjuvant hormone therapy with brachytherapy is controversial. It is used to<br />

reduce the <strong>prostate</strong> volume when it exceeds 50 ml, in order to facilitate brachytherapy. Volume<br />

reduction decreases the total isotope activity required, potentially improves implant dosimetry<br />

and decreases pubic arch interference. 70<br />

Patient selection (exclusions)<br />

• Prostate size >50 ml<br />

• Recent TURP<br />

• Significant urinary outflow obstruction<br />

• Previous AP resection<br />

• Previous high dose pelvic radiotherapy<br />

27


Side-effects<br />

• A review of 16 studies by Crook et al. showed acute adverse events as: 67<br />

o Irritant urinary symptoms: 46−54%<br />

o Acute urinary retention: 1−14%<br />

o Acute proctitis: 1−2%<br />

o Chronic adverse events (rein<strong>for</strong>ced by Wills et al., 1999 71 ):<br />

• Incontinence: 5−6%<br />

• Haematuria: 1−2%<br />

• Strictures: 1−2%<br />

• Proctitis: 1−3%<br />

• Erectile dysfunction: 4−14% (or up to 38% in Wills et al., 1999 71 and up to 50% at 5 years<br />

in Merrick et al., 2001 68 ).<br />

Clinical evidence<br />

Table 4: Experience using trans-perineal implants as monotherapy (T1, T2 <strong>cancer</strong>s)<br />

Author Number of men Isotope<br />

*Series excluded Gleason grade >7, 38% with pre-treatment PSA ≤4.0 ng/ml<br />

Median follow-up PSA results<br />

(months) (months)<br />

Beyer & Priestley,<br />

79% with PSA<br />

489 I-125 35<br />

199772


Novel therapies<br />

Cryotherapy/High-intensity focused ultrasonography (HIFU)<br />

• The development of third-generation <strong>prostate</strong> cryotherapy has allowed the introduction of<br />

ultra-thin needles to deliver a minimally-invasive treatment <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong> patients in the<br />

primary and salvage setting<br />

79, 80<br />

o Early results established the low incidence of serious side-effects and good PSA control<br />

o Longer-term follow-up series show biochemical DFS at 10 years of 80.56% <strong>for</strong> low-risk,<br />

74.16% <strong>for</strong> moderate-risk and 45.54% <strong>for</strong> high-risk <strong>prostate</strong> <strong>cancer</strong> patients<br />

• This treatment has been approved by the American Urological Association and the European<br />

Association of Urology <strong>for</strong> treatment of patients with primary and radiation-failed <strong>prostate</strong><br />

<strong>cancer</strong><br />

• In the NICE guidelines, the minimally-invasive treatments of cryosurgery and HIFU were<br />

considered to be experimental and <strong>for</strong> use only within the clinical trial setting 29<br />

o After further clarification, it was agreed that these treatments would be funded by Primary<br />

Care Trusts provided patient data were collected in an approved way. Following consultation<br />

with the BAUS section of Oncology, a national database has been established to collect data<br />

from <strong>prostate</strong> <strong>cancer</strong> patients undergoing cryotherapy or HIFU <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong>. Inclusion<br />

of patients in this database is considered necessary to fulfil the NICE guidelines.<br />

• These treatments may be offered to patients with stage T1, T2 or T3 as a primary treatment or in<br />

salvage <strong>prostate</strong> <strong>cancer</strong> patients.<br />

o The staging requirements are a positive biopsy and MRI and bone scan showing no evidence<br />

of metastatic spread.<br />

29


Locally Advanced Disease (Non-metastatic):<br />

Management Options<br />

Figure 3: Treatment algorithm <strong>for</strong> locally advanced (non-metastatic) disease<br />

Key Questions <strong>for</strong> the <strong>MDT</strong><br />

• TNM stage?<br />

• Gleason grade?<br />

• PSA/PSA Kinetics?<br />

• Age/co-morbidity/life<br />

expectancy?<br />

• Symptoms:<br />

– Bowel?<br />

– Urine IPSS score?<br />

• Sexual function?<br />

• Family history?<br />

• Bone pain?<br />

• Osteoporosis risk?<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

High-risk localised<br />

<strong>MDT</strong><br />

Diagnostic Tests<br />

• Digital rectal exam (DRE)<br />

• PSA<br />

• Transrectal ultrasound<br />

(TRUS)/biopsy<br />

• MRI/CT pelvic scan<br />

• Bone scan<br />

• Consider lymph node sampling<br />

(if this will determine changes<br />

in management approach)<br />

MANAGEMENT OPTIONS 1<br />

T1/T2 N+ Mo<br />

T3/T4 No/N+ Mo<br />

External beam radiotherapy (EBRT)<br />

+/- HDR brachytherapy boost<br />

+/-neo-adjuvant, concomitant<br />

and adjuvant hormone therapy<br />

Hormone therapy alone<br />

Watchful waiting<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

Key Discussion Points<br />

with the Patient<br />

• Survival/prognosis?<br />

• Treatment options?<br />

• Treatment side-effects?<br />

• Impact on quality of life?<br />

• Importance of:<br />

– Psychological impact<br />

to patient and family?<br />

– Family history?<br />

– Clinical trials?<br />

– Sexual function?<br />

– Urinary function?<br />

– Bowel function?<br />

– Physical strength, energy?<br />

– Level of activity?<br />

– Accessibility to Rx?<br />

30


The term locally advanced <strong>prostate</strong> <strong>cancer</strong> can be used to encompass a spectrum of disease profiles<br />

that may include any of the following:<br />

• Clinical stage T3, T4 or N1 <strong>cancer</strong>s without evidence of distant metastases (M0)<br />

• Clinical stages T1 and T2 (‘localised’) at diagnosis, where ‘high-risk’ features (PSA concentration<br />

≥20 ng/ml or Gleason grade ≥8) indicate the likelihood of extraprostatic invasion or clinically<br />

undetectable metastatic disease<br />

• Pathological stage pT2 or pT3 disease with ‘high-risk’ features due to upstaging from additional<br />

pathological in<strong>for</strong>mation after radical <strong>prostate</strong>ctomy<br />

Men with locally advanced or high-risk <strong>prostate</strong> <strong>cancer</strong> generally have a significant risk of disease<br />

progression and <strong>cancer</strong>-related death if left untreated. These patients present two specific challenges.<br />

There is a need <strong>for</strong> local control and also a need to treat any microscopic metastases likely to be<br />

present but undetectable until disease progression. The optimal treatment approach will often<br />

there<strong>for</strong>e utilise <strong>multi</strong>ple modalities. The exact combinations, timing and intensity of treatment<br />

continue to be strongly debated. Management decisions should be made after all treatments have<br />

been discussed by the <strong>MDT</strong> and the balance of benefits and side-effects of each therapy modality have<br />

been considered by the patient with regard to their own individual circumstances.<br />

Watchful waiting<br />

Overview<br />

• Data on the use of watchful waiting in locally advanced disease is sparse.<br />

• Two non-randomised studies concluded that immediate orchidectomy was not associated with a<br />

81, 82<br />

survival advantage compared with therapy delayed until metastatic progression.<br />

• A pooled analysis of data from 2 RCTs involving 1036 men with locally advanced disease not<br />

suitable <strong>for</strong> curative treatment (T2−T4) suggested no survival benefit <strong>for</strong> immediate versus<br />

delayed hormone therapy at 1, 5 or 10 years. 83<br />

Clinical evidence<br />

• Adolfsson et al. prospectively followed 50 patients with locally advanced <strong>prostate</strong> <strong>cancer</strong> who<br />

were only treated upon patient request or when they became symptomatic. All patients were<br />

followed-up <strong>for</strong> more than 144 months, or had died be<strong>for</strong>e that point. OS and DFS at 5, 10 and 12<br />

years was 68% and 90%, 34% and 74%, and 26% and 70%, respectively. 84<br />

31


Hormone therapy<br />

Immediate versus deferred hormonal treatment<br />

• Immediate versus deferred treatment <strong>for</strong> advanced <strong>prostate</strong> <strong>cancer</strong> was investigated by the MRC<br />

Prostate Working Party Investigators Group. An RCT of 943 men with asymptomatic metastases or<br />

locally advanced disease, not suitable <strong>for</strong> curative treatment, was undertaken, with randomisation<br />

to immediate or deferred hormone therapy. 85<br />

o There was a significant advantage in the immediate treatment group in terms of distant<br />

progression. Mortality was only significantly changed by treating immediately in those with<br />

M0 disease (Table 5).<br />

o A modest but statistically significant increase in OS was seen in the immediate treatment<br />

group, but not significant difference in <strong>prostate</strong> <strong>cancer</strong> mortality or symptom-free survival<br />

was demonstrated.<br />

o Due consideration must there<strong>for</strong>e be given to potential effects of long-term ADT versus the<br />

potential avoidance of such effects in patients if hormone therapy is deferred. 86<br />

Table 5: Effect of immediate versus deferred hormonal treatment 85<br />

Immediate Deferred<br />

Distant progression 26% 45%<br />

Mortality due to <strong>prostate</strong> <strong>cancer</strong> M0 disease 31.6% 48.8%<br />

M1 disease No significant No significant<br />

difference difference<br />

Hormone therapy plus standard care<br />

• The <strong>multi</strong>centre, international Early Prostate Cancer (EPC) study evaluated the efficacy and<br />

tolerability of adding the non-steroidal anti-androgen bicalutamide 150 mg once-daily to<br />

standard care (<strong>prostate</strong>ctomy, radiotherapy or watchful waiting). 8113 patients with localised or<br />

locally advanced non-metastatic <strong>prostate</strong> <strong>cancer</strong> were included. 87<br />

o Objective PFS and OS were defined as the primary endpoints. At a third analysis, the median<br />

follow-up was 7.4 years. Exploratory analyses were also conducted to determine the efficacy<br />

of bicalutamide in clinically relevant subgroups.<br />

o A trend towards increased survival was seen <strong>for</strong> the subgroup treated with bicalutamide who<br />

would otherwise have undergone watchful waiting (HR 0.81; 95%CI: 0.66−1.01; p=0.06).<br />

o A significant improvement in objective PFS in favour of bicalutamide 150 mg <strong>for</strong> all locally<br />

advanced disease patients was demonstrated irrespective of standard care received (HR<br />

0.69; 95%CI: 0.58−0.83; p


Radiotherapy plus hormone therapy<br />

• A study by Widmark et al. has shown that the addition of radiotherapy to hormone therapy <strong>for</strong><br />

men with locally advanced or high-risk <strong>prostate</strong> <strong>cancer</strong> halves the 10-year <strong>prostate</strong> <strong>cancer</strong>-specific<br />

mortality and substantially decreases overall mortality. 88<br />

o This phase III study comparing endocrine therapy with and without local radiotherapy<br />

randomised 875 patients with locally advanced <strong>prostate</strong> <strong>cancer</strong> (T3; 78%; PSA concentration<br />


EBRT +/− neoadjuvant, concomitant and adjuvant hormone therapy<br />

Radiotherapy alone<br />

• In locally advanced disease, EBRT alone has been shown to have a poorer outcome than in<br />

localised <strong>prostate</strong> <strong>cancer</strong>. Consequently, combination therapy with radiotherapy and hormone<br />

therapy is rapidly being accepted as standard practice.<br />

• Although it has been widely used, there are still many uncertainties associated with radical<br />

radiotherapy with regard to the optimum dose and field size (particularly to what extent the<br />

treatment volume should try to include pelvic lymph nodes). These uncertainties are compounded<br />

by the availability of new approaches to the delivery of radiotherapy, such as IMRT. The latter is<br />

intended to shape the radiation field more precisely to the tumour volume, thereby reducing the<br />

side-effects of treatment and possibly allowing dose escalation that enhances its local efficacy.<br />

Radiotherapy target volume/lymph nodes<br />

• In high-risk patients the consensus is that the seminal vesicles should be included.<br />

• The RTOG 9413 trial was designed to determine whether there was an advantage in terms<br />

of PFS with total androgen suppression, whole pelvic radiotherapy followed by a <strong>prostate</strong><br />

boost compared with total androgen suppression and <strong>prostate</strong>-only radiotherapy. The trial<br />

also investigated the timing of hormone therapy with a further randomisation. One group<br />

received neoadjuvant hormone therapy followed by concurrent total androgen suppression and<br />

radiotherapy while the other group was treated with radiotherapy followed by adjuvant hormone<br />

therapy. Patients with non-metastatic disease but an estimated risk of lymph node involvement of<br />

>15% were randomised between the 4 arms. 89<br />

o The difference in OS <strong>for</strong> the 4 arms was statistically significant (p=0.027).<br />

o However, no statistically significant differences were found in PFS or OS between neoadjuvant<br />

versus adjuvant hormone therapy and whole pelvis radiotherapy compared with <strong>prostate</strong>only<br />

radiotherapy. A trend towards a difference was found in PFS (p=0.065) in favour of the<br />

whole pelvic radiotherapy + neoadjuvant hormone arm compared with the <strong>prostate</strong>-only<br />

radiotherapy + neoadjuvant hormones and whole pelvic radiotherapy + adjuvant hormone<br />

treatment arms.<br />

o These results have demonstrated that when neoadjuvant hormone therapy is used in<br />

conjunction with radiotherapy, whole pelvic treatment yields a better PFS than <strong>prostate</strong>-only<br />

radiotherapy. It also showed an improved OS when whole pelvic radiotherapy was combined<br />

with neoadjuvant rather than short-term adjuvant hormone therapy.<br />

Radiotherapy dose escalation<br />

• Evidence suggests that patients treated with radiotherapy to the <strong>prostate</strong> have a significantly<br />

better outcome, because the dose to the gland is increased. The benefit is greatest in those<br />

patients with high-risk features.<br />

• Debate remains over the best way of increasing the dose without significantly increasing normal<br />

tissue toxicity. 3D-CRT, IMRT and high dose rate (HDR) brachytherapy boost are methods currently<br />

under evaluation.<br />

34


• Evidence of the benefits of dose escalation has been demonstrated <strong>for</strong> T1−T3 <strong>prostate</strong> <strong>cancer</strong> by<br />

Pollack in a phase III randomised study at the MD Anderson Hospital. 53<br />

o A total of 305 men were randomised between 1993 and 1998 to compare the efficacy of<br />

70 Gy versus 78 Gy with a median follow-up of 60 months. The primary endpoint was FFF,<br />

including biochemical failure, which was defined as three rises in PSA level.<br />

o The FFF rates <strong>for</strong> the 70 Gy and 78 Gy arms at 6 years were 64% and 70%, respectively<br />

(p=0.03).<br />

o Dose escalation to 78 Gy preferentially benefited those with a pre-treatment PSA<br />

concentration >10 ng/ml; the FFF rate was 62% <strong>for</strong> the 78 Gy arm versus 43% <strong>for</strong> those who<br />

received 70 Gy (p=0.01). For patients with a pre-treatment PSA concentration ≤10 ng/ml, no<br />

significant dose response was found, with an average 6-year FFF rate of about 75%.<br />

o Although no difference in OS was observed, the freedom from distant metastasis rate was<br />

higher <strong>for</strong> those with PSA concentrations >10 ng/ml who were treated to 78 Gy (98% versus<br />

88% at 6 years; p=0.056).<br />

• Dearnaley and colleagues have reported their findings from the MRC RT01 study. 54<br />

o In this 3D-CRT trial 843 men were randomised to standard dose of 64 Gy compared to an<br />

escalated dose of 74 Gy with all men receiving neoadjuvant hormone therapy.<br />

o Patients receiving the conventional dose had 5-year biochemical PFS rates of 60% compared<br />

with 71% in the dose-escalated arm. Advantages were also seen in terms of PFS and the<br />

decreased use of androgen suppression.<br />

o Although these and other studies have shown benefits from dose escalation with 3D-CRT<br />

techniques, this has been offset to a degree by a reported increase in late rectal toxicity.<br />

• Prospective non-randomised studies at the Memorial Sloan Kettering <strong>cancer</strong> centre have<br />

compared the outcomes of 1100 men who received doses in the range of 64 to 70 Gy and 76 to<br />

86 Gy using IMRT. The results were evaluated within prognostic risk groups (using clinical stage,<br />

Gleason grade and presenting PSA). 55<br />

• They demonstrated that increasing the dose delivered beyond 70.2 Gy in men with intermediate-<br />

and high-risk disease improved the 5-year actuarial PSA relapse-free survivals from 50% to 70%<br />

and 21% to 47%, respectively, in these two risk categories.<br />

• These results confirmed the advantage of dose escalated radiotherapy with doses of over 80 Gy.<br />

IMRT has the potential to reduce late rectal toxicity as shown in a further study by Zelefsky which<br />

reports 3-year actuarial ≥ grade 2 gastrointestinal toxicity at 4%. 56<br />

• A further development under investigation involves a change in the traditional fractionation<br />

schedules. Hypofractionation may improve <strong>cancer</strong> control <strong>for</strong> the same level of radiation-related<br />

toxicity and be a more effective treatment <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong> with a predicted low alpha/<br />

beta ratio. Phase II dose escalation studies using shortened schedules of hypofractionated IMRT<br />

regimens have indicated acceptable early toxicity. 57<br />

• The CHHiP study is currently recruiting patients in the UK to compare standard fractionation IMRT<br />

(74 Gy in 37 fractions) to two hypofractionated IMRT regimens (60 Gy in 20 fractions or 57 Gy in<br />

19 fractions) in combination with neoadjuvant hormone therapy. 58<br />

35


HDR brachytherapy boost<br />

• HDR brachytherapy is a safe, reproducible and effective way of boosting conventional EBRT. There<br />

is published evidence <strong>for</strong> this approach demonstrating improved biochemical control and causespecific<br />

survival without a significant increase in toxicity.<br />

• Currently, HDR brachytherapy is used in combination with EBRT and is felt appropriate <strong>for</strong><br />

patients with high-risk localised or locally advanced <strong>prostate</strong> <strong>cancer</strong>. 90<br />

EBRT plus neoadjuvant hormone therapy<br />

• Neoadjuvant hormone therapy reduces <strong>prostate</strong> volume by 30−40%. 59, 60 This can reduce the size<br />

of the treatment field and as a result the level of toxicity experienced.<br />

• There are also reports of an additive or synergistic effect on tumour cell kill with combined<br />

therapy. Theories as to the mechanism of this include improved oxygenation by reducing tumour<br />

bulk and movement of hormone-responsive cells into a resting phase, which could reduce<br />

repopulation rate and enhance tumour cell death (increased apoptosis). 61<br />

• The RTOG 86-10 trial randomised 471 men with T2−T4 <strong>prostate</strong> <strong>cancer</strong> to radiotherapy +/− 4<br />

months of ADT (goserelin 3.6 mg depot once-monthly plus flutamide 250mg tid) be<strong>for</strong>e and<br />

during EBRT or to radiotherapy alone. The median follow-up was 6.7 years <strong>for</strong> all patients and 8.6<br />

years <strong>for</strong> surviving patients. 62<br />

o At median follow-up of 8.7 years <strong>for</strong> surviving patients, there was a trend to improved<br />

survival (8-year survival 53% versus 44%, p=0.1) <strong>for</strong> those treated by hormone therapy with<br />

radiotherapy, which was significant <strong>for</strong> the subgroup with Gleason grade 2−6 disease (70%<br />

versus 52%, p=0.015). 62<br />

o Ten-year OS estimates (43% versus 34%) and median survival times (8.7 versus 7.3 years)<br />

favoured combined therapy with hormones and radiation compared to radiation treatment<br />

alone; however, these differences did not reach statistical significance (p=0.12). 63<br />

o There was a statistically significant improvement in 10-year disease-specific mortality<br />

(23% versus 36%; p=0.01), distant metastases (35% versus 47%; p=0.006), DFS (11% versus<br />

3%; p


Clinical evidence<br />

• Adjuvant androgen suppression immediately after radical radiotherapy has been shown to<br />

significantly increase OS, PFS, and significantly reduce local progression, distant metastases and<br />

biochemical progression in several large randomised studies.<br />

• Bolla et al. (EORTC 22863) randomised 415 patients with locally advanced non-metastatic <strong>prostate</strong><br />

<strong>cancer</strong> (T1−4, Nx, M0) to receive either radiotherapy with immediate goserelin 3.6 mg therapy<br />

(every 4 weeks <strong>for</strong> 3 years) plus cyproterone acetate (CPA) during the first month of treatment <strong>for</strong><br />

disease flare (n=207) or radiotherapy alone (n=208). 91<br />

o After a mean follow-up of 66 months, the 5-year DFS rate was 74% versus 40% <strong>for</strong> the<br />

combined and EBRT alone groups, respectively (p=0.001).<br />

o Furthermore, the corresponding OS and <strong>cancer</strong>-specific survival rates at 5 years were 78% and<br />

62% (p=0.0002), while the <strong>cancer</strong>-specific survival rates were 94% versus 79%.<br />

o A further analysis of this study with a median follow-up of 9.1 years reported 192 deaths<br />

(112 in radiotherapy alone and 80 in the combined treatment arm). 92<br />

o The addition of 3 years of ADT increased the 10-year OS from 39.8% to 58.1% (HR 0.60;<br />

95%CI: 0.45−0.80; p=0.0004), clinical PFS from 22.7% to 47.7% (HR 0.42; 95%CI: 0.33−0.55;<br />

p


• Hanks et al. (RTOG 92-02) investigated the use of long-term androgen suppression following<br />

neoadjuvant hormonal cytoreduction and radiotherapy in locally advanced <strong>prostate</strong> <strong>cancer</strong><br />

(T2c to T4 with no extra pelvic lymph node involvement and PSA


Locally Advanced (Non-metastatic) Disease:<br />

Relapse after Primary Treatment<br />

Figure 3: Treatment algorithm <strong>for</strong> locally advanced (non-metastatic) disease (cont.)<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

Post-radical <strong>prostate</strong>ctomy<br />

MANAGEMENT OPTIONS 2<br />

Failed local Rx with rising PSA<br />

and/or local recurrence+ Mo<br />

Active surveillance/watchful waiting Active surveillance/watchful waiting<br />

Hormone therapy alone Hormone therapy alone<br />

External beam radiotherapy<br />

(EBRT) +/– concomitant +/–<br />

adjuvant hormone therapy<br />

Post-radical radiotherapy/<br />

brachytherapy<br />

Salvage <strong>prostate</strong>ctomy<br />

Novel local salvage therapy<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

Rising PSA levels<br />

• The PSA concentration at which to define treatment failure after <strong>prostate</strong>ctomy varies<br />

in the literature.<br />

Definitions of recurrence<br />

• The Phoenix definition of relapse after radiotherapy is nadir plus 2. 97<br />

• Patients whose PSA never falls to an undetectable level in the post-operative period are generally<br />

considered to have systemic disease. However, some may have local disease amenable to salvage<br />

radiotherapy, and so need to be assessed to determine the best management plan.<br />

• A PSA concentration that rises rapidly in the post-operative setting may be indicative of metastatic<br />

disease, while a PSA that remains undetectable over a long period then gradually rises may be<br />

more likely to indicate local recurrence.<br />

39


• Pound et al. carried out a retrospective review of 1997 men undergoing radical <strong>prostate</strong>ctomy<br />

by a single surgeon <strong>for</strong> clinically localised disease with no neoadjuvant or adjuvant treatment. 98<br />

A PSA ≥0.2 ng/ml was deemed evidence of recurrence.<br />

o At 15 years, 15% had PSA elevation and 34% of these had developed metastases.<br />

o The median time from PSA elevation to metastatic disease was 8 years.<br />

o After development of metastases, the median actuarial time to death was 5 years. In the<br />

survival analysis, time to biochemical progression, Gleason grade and PSA doubling time<br />

were predictive of the probability and time to the development of metastatic disease.<br />

• After completion of radiotherapy and hormonal treatment, testosterone recovery usually occurs.<br />

This may cause some PSA elevation that is related to normal <strong>prostate</strong> tissue recovery and not<br />

disease recurrence.<br />

• The definition of disease recurrence in the setting of combined therapy remains a matter<br />

of debate and consensus is awaited.<br />

• Benign PSA rises (PSA bounce) occur in approximately 12% of patients following EBRT<br />

and 30% following LDR brachytherapy in the absence of neoadjuvant hormonal treatment<br />

(starting between 18 months and 2 years after treatment).<br />

Local recurrence after radical <strong>prostate</strong>ctomy<br />

Overview<br />

• Overall, approximately 40% of patients who have a radical <strong>prostate</strong>ctomy have biochemical<br />

evidence of recurrence at some point.<br />

• Determining whether relapse is local or distant is important in determining optimal treatment.<br />

However, post-<strong>prostate</strong>ctomy imaging is unhelpful as this will always be negative, so there is<br />

no way of defining local versus distant disease categorically. Other factors that may aid this<br />

distinction include:<br />

o Timing and pattern of PSA relapse (rapid rise post-operatively indicates distant spread)<br />

o Involvement of seminal vesicles or lymph nodes<br />

o Margin status at surgery<br />

o Gleason grade<br />

• Radical salvage treatment is usually via radiotherapy to the <strong>prostate</strong> bed +/− hormone therapy.<br />

The optimal time of treatment, i.e. immediate adjuvant or early salvage EBRT, is currently<br />

uncertain. The timing and duration of hormone therapy is also unclear.<br />

• The RADICALS study is investigating the timing of radiotherapy (immediate versus early salvage)<br />

and hormone duration. 50<br />

40


Clinical evidence<br />

• The EORTC 22911 study was designed to investigate benefit <strong>for</strong> immediate post-operative<br />

radiotherapy in 502 patients with pT3 disease or positive surgical margins as opposed to<br />

radiotherapy offered <strong>for</strong> biochemical or clinical relapse. 48<br />

o After a median follow-up of 5 years, biochemical PFS was significantly improved in the<br />

immediate radiotherapy group (74.0% versus 52.6%; p


Recurrence after radical radiotherapy<br />

Overview<br />

• The therapeutic options <strong>for</strong> recurrence following radiotherapy include:<br />

o Salvage radical <strong>prostate</strong>ctomy: associated with 5-year biochemical DFS rates of 55−69%, but<br />

the technique is associated with a significant incidence of complications, such as rectal injury,<br />

anastamotic stricture and urinary incontinence.<br />

o Salvage cryotherapy: 5-year biochemical PFS ranges from 40% to 73%. The complications<br />

of salvage cryotherapy are erectile dysfunction, pelvic, rectal or perineal pain, recto-urethral<br />

fistula, bladder outlet obstruction and urethral stricture.<br />

o Salvage HIFU is currently under investigation.<br />

o Hormone therapy can be given in combination with local treatments or as monotherapy.<br />

Clinical evidence<br />

• Touma et al. reviewed the efficacy and safety of salvage radical <strong>prostate</strong>ctomy, cryotherapy and<br />

brachytherapy <strong>for</strong> recurrence following definitive radiotherapy. 100<br />

o Salvage radical <strong>prostate</strong>ctomy is associated with 5-year biochemical DFS rates of 55−69%, but<br />

the technique is associated with a significant incidence of complications, such as rectal injury,<br />

anastomotic stricture and urinary incontinence.<br />

o The four studies of salvage cryotherapy reviewed used varying definitions of recurrence.<br />

The 5-year biochemical PFS ranged from 40% when failure was defined as PSA 2 above<br />

nadir, to 62% and 73% when failure was defined as PSA greater than 2 and greater than 4,<br />

respectively.<br />

o The complications of salvage cryotherapy are erectile dysfunction, pelvic, rectal or perineal<br />

pain, rectourethral fistula, bladder outlet obstruction and urethral stricture.<br />

• In one trial of 49 patients reported by Grado et al., complications included lower urinary tract<br />

symptoms, especially in the first 3 months. TURP was necessary in 14% of patients due to bladder<br />

outlet obstruction, while haematuria occurred in 4% of cases, stricture in 6%, rectal ulcers in 4%<br />

and rectal bleeding necessitating colostomy in 2%. 101<br />

• In 71 patients with localised disease following EBRT, following HIFU, 80% demonstrated negative<br />

biopsies and 61% had a nadir PSA concentration


Advanced Prostate Cancer (Metastatic): Management Options<br />

Figure 4: Treatment algorithm <strong>for</strong> advanced (metastatic) disease<br />

Key Questions <strong>for</strong> the <strong>MDT</strong><br />

• TNM stage?<br />

• Gleason grade?<br />

• PSA/PSA Kinetics?<br />

• Age/co-morbidity/life<br />

expectancy?<br />

• Symptoms:<br />

– Bowel?<br />

– Urine IPSS score?<br />

• Sexual function?<br />

• Family history?<br />

• Bone pain?<br />

• Osteoporosis risk?<br />

• Palliative care referral?<br />

Is this patient suitable <strong>for</strong> clinical trial?<br />

Palliative Care<br />

• Pain control<br />

• Local radiotherapy<br />

Bone pain<br />

Spinal cord compression<br />

Nerve root compression<br />

• Continual assessment<br />

Second-/third-line if<br />

androgen sensitive?<br />

<strong>MDT</strong><br />

Diagnostic Tests<br />

• Digital rectal exam (DRE)<br />

• PSA<br />

• Transrectal ultrasound (TRUS)/<br />

biopsy (non mandatory if PSA<br />

>100 and radiological evidence<br />

of metastases)<br />

• Biochemistry screen<br />

• Full blood count<br />

• Bone scan<br />

• MRI/CT pelvic scan<br />

MANAGEMENT OPTIONS<br />

Proven M1/M2<br />

First line hormone therapy<br />

Significant rise in PSA Clinical/<br />

radiological progression<br />

Observation<br />

Second-/third-line hormone<br />

therapy<br />

Further progression<br />

Ongoing Support<br />

Local patient support network<br />

Role of nurse/GP/healthcare professional <strong>team</strong><br />

Key Discussion Points<br />

with the Patient<br />

• Survival/prognosis?<br />

• Treatment options?<br />

• Treatment side-effects?<br />

• Impact on quality of life?<br />

• Importance of:<br />

– Psychological impact<br />

to patient and family?<br />

– Family history?<br />

– Clinical trials?<br />

– Sexual function?<br />

– Urinary function?<br />

– Bowel function?<br />

– Physical strength, energy?<br />

– Level of activity?<br />

– Accessibility to Rx?<br />

HRPC<br />

Based on MRC evidence, the majority of patients with metastatic disease should be treated.<br />

Deferred treatment is acceptable only in highly selected, in<strong>for</strong>med patients.<br />

Clinical trials<br />

Chemotherapy<br />

Strontium<br />

Bisphosphonate<br />

43


Primary hormone therapy<br />

Overview<br />

• ADT is standard first-line treatment <strong>for</strong> the management of patients with metastatic disease.<br />

ADT can involve orchidectomy, LHRH agonists, anti-androgens and gonadotrophin-releasing<br />

hormone (GnRH) antagonists.<br />

• Orchidectomy remains the gold-standard ADT against which all other treatments are compared<br />

because of its rapid effects on total testosterone concentrations. 103<br />

104, 105<br />

• Survival appears equivalent with LHRH agonists and orchidectomy.<br />

• A meta-analysis has indicated that 2-year survival may be worse with medical treatment than<br />

with orchidectomy. 106<br />

• Patients, however, prefer medical treatment and in terms of usage, drug treatment represents<br />

the standard of care at all disease stages. 107−109<br />

• GnRH antagonists are clinically equivalent to LHRH agonists without causing the initial<br />

testosterone surge seen with LHRH agonists. Now licensed on the evidence of phase III clinical<br />

trial data, degarelix demonstrates reduced testosterone concentrations to below castrate levels<br />

in 3 days (90% decrease in median testosterone compared with leuprolide group experiencing<br />

a 65% increase in median testosterone levels; p


Table 6: Effect of immediate versus deferred hormonal treatment 85<br />

Combined (maximum) androgen blockade<br />

• There is debate over the use of combined androgen blockade (CAB). In 2000, the Prostate Cancer<br />

Trialists’ Collaborative Group published a meta-analysis of the available trials of CAB versus<br />

monotherapy. The analysis included 27 trials, which incorporated 8275 men, representing 98%<br />

111, 112<br />

of men ever randomised in trials of CAB versus monotherapy.<br />

o The 5-year survival <strong>for</strong> all patients receiving CAB was 25.4%, compared with 23.6% <strong>for</strong><br />

patients receiving monotherapy.<br />

o In subgroup analyses, patients treated with CPA seemed to fare slightly worse than those<br />

treated with flutamide or nilutamide, mostly secondary to non-<strong>prostate</strong> <strong>cancer</strong>-related<br />

deaths.<br />

• If the CPA studies were excluded, the results were as follows: 111<br />

Immediate Deferred<br />

Distant progression 26% 45%<br />

Mortality due to <strong>prostate</strong> <strong>cancer</strong> M0 disease 31.6% 48.8%<br />

M1 disease No significant No significant<br />

difference difference<br />

o CAB with flutamide alone was associated with an 8% reduction in the risk of death (95%CI:<br />

0.86−0.98; p=0.02), which translates to a small but significant improvement in 5-year survival<br />

over castration alone.<br />

o CAB with flutamide plus nilutamide was associated with an 8% reduction in the risk of death<br />

(95%CI: 1.00−1.27; p=0.005), which translates to a small but significant improvement in 5-year<br />

survival of 2.9% over castration alone.<br />

o Conversely, CAB with CPA is associated with an increased risk of death of 13% (95%CI:<br />

1.00−1.27; p=0.04), which translates to a small but significant reduction in 5-year survival<br />

of 2.8% over castration alone.<br />

• It can be concluded that the choice of anti-androgen used <strong>for</strong> CAB has an impact on outcome, and<br />

that CAB with a non-steroidal anti-androgen may offer a small but significant survival benefit.<br />

45


Second- or third-line hormone therapy<br />

• Some patients will respond to second-line hormone therapy (CAB with the addition of an antiandrogen,<br />

anti-androgen withdrawal, diethylstilboestrol [DES]). Anti-androgen withdrawal<br />

responses are seen in approximately 25% of cases who have been treated with first-line CAB<br />

or have had substantial (>1 year response) to second-line CAB.<br />

• A common second-line treatment is the addition of an anti-androgen. A retrospective analysis<br />

of 122 patients who received the addition of bicalutamide 50 mg to goserelin <strong>for</strong> PSA and clinical<br />

progression showed a >50% decrease in PSA concentration in 30% of patients (responders) and<br />

a reduction in PSA concentration in 75% of all patients. The median duration of response from<br />

start of bicalutamide 50 mg was 291 days <strong>for</strong> responders and 193 days <strong>for</strong> the population as a<br />

whole. Those patients with a short duration of response to goserelin monotherapy (


Chemotherapy, Strontium and Bisphosphonates<br />

Those patients who do not respond to hormone therapy are considered to have hormone-refractory<br />

<strong>prostate</strong> <strong>cancer</strong> (HRPC; i.e. unresponsive to all hormone therapies) or castrate-refractory <strong>prostate</strong><br />

<strong>cancer</strong> (unresponsive to treatment with LHRH agonists) and are candidates <strong>for</strong> chemotherapy, novel<br />

therapies and/or symptomatic local treatments.<br />

Chemotherapy<br />

• A prospective study by Tannock in 1996 compared the benefits of mitoxantrone 12 mg/m² every<br />

3 weeks plus prednisone 5 mg twice-daily with prednisone alone in 161 men with symptomatic<br />

HRPC. 116<br />

o The primary endpoint was palliative response defined as a 2-point decrease in pain as assessed<br />

by a 6-point pain scale.<br />

o There was a significant advantage to the chemotherapy combination with a 29% pain<br />

response compared to 12% with steroids alone.<br />

o The duration of palliation was 43 weeks versus 18 weeks (p


Side-effects<br />

• Side-effects of chemotherapy depend on the treatment, but usually include fatigue, nausea and<br />

vomiting, diarrhoea, hair loss and increased susceptibility to infection. Specific therapies to handle<br />

these side-effects may be necessary to improve the patient’s quality of life.<br />

Strontium<br />

• Metastatic pain can be palliated effectively with systemic radionuclide therapy with strontium<br />

chloride.<br />

• Relief of bone pain starts within 2 weeks. Possible initial bone pain flare may occur within 2 days,<br />

lasting 2−4 days.<br />

o Pain relief lasts 4−15 months.<br />

o 75−80% of patients experience significant palliation of pain.<br />

• A Canadian collaborative study showed significant improvement in quality of life, increased time<br />

to further metastases, significant reduction in the amount of additional radiotherapy needed,<br />

and significant falls in PSA and alkaline phosphatase. 120<br />

• Strontium is not associated with improvements in OS. 121<br />

• Four randomised clinical trials have reviewed the use of strontium. 122<br />

o One trial reported significant improvement in pain control, two trials reported fewer new<br />

sites of pain.<br />

o One trial showed no significant difference in pain control compared to a placebo but an<br />

improved 2-year survival rate.<br />

• A randomised clinical trial examining strontium versus placebo found a significant increase in<br />

median time to progression, but no significant effects on median OS or clinical response. 123<br />

Side-effects<br />

• The most notable side-effect of strontium is mild haematological suppression with a fall in<br />

circulating platelet and leukocyte counts recognised in most patients.<br />

o With usual therapeutic doses, platelets typically fall by 30% and leucocytes by 20%.<br />

o Clinically significant toxicity is rare, but its use is not recommended in patients with severely<br />

compromised bone marrow, platelet count


Bisphosphonates<br />

• The benefits of zoledronic acid, in combination with hormone therapy have been investigated<br />

in a study by Saad in men with HRPC and bone metastases. 124 This was a <strong>multi</strong>centre, randomised,<br />

placebo-controlled trial evaluating the efficacy of zoledronic acid 4 mg administered every<br />

3 weeks in 422 patients with HRPC <strong>for</strong> 15 months, with an option to continue <strong>for</strong> an additional<br />

9 months.<br />

o At the 2-year analysis, treatment with zoledronic acid was found to significantly reduce the<br />

percentage of patients with at least one skeletal-related event (SRE; defined as radiation <strong>for</strong><br />

bone pain or to prevent pathological fracture/spinal cord compression; pathological fracture;<br />

spinal cord compression; surgery to bone; change in antineoplastic therapy) compared with<br />

placebo (38% versus 49%; p=0.028). All SREs were delayed.<br />

o Zoledronic acid also significantly delayed the time to first SRE by around 6 months (median<br />

488 versus 321 days; p=0.009). Furthermore, patients in the zoledronic acid group had<br />

consistently lower incidences of all types of SRE than the placebo group. Pain scores were<br />

consistently lower in patients taking zoledronic acid 4mg than placebo, and significantly<br />

at 3, 9, 18, 21 and 24 months (p


Palliative Care<br />

Overview<br />

• Radiotherapy has been a mainstay in the palliation of painful metastatic bone lesions. Palliative<br />

radiotherapy can also aid other complications of metastatic disease, such as compression of the<br />

spinal cord or a nerve root, haematuria, ureteric obstruction, perineal discom<strong>for</strong>t caused by the<br />

local progression of <strong>prostate</strong> <strong>cancer</strong>, and symptomatic metastatic lymphadenopathy.<br />

Clinical evidence<br />

• Good evidence <strong>for</strong> the role of radiotherapy in palliation comes from McQuay et al. This systematic<br />

review covered 20 trials, which reported on 43 different radiotherapy fractionation schedules, and<br />

eight studies of radioisotopes. 128<br />

o Radiotherapy produced complete pain relief at 1 month in 395 out of 1580 (25%) patients,<br />

and at least 50% relief in 788 out of 1933 (41%) patients at some time during the trials.<br />

o In the largest trial, which included 759 patients, 52% achieved complete pain relief within<br />

4 weeks and the median duration of complete relief was 12 weeks.<br />

o The study found no difference between the use of radioisotopes (such as strontium) and<br />

EBRT <strong>for</strong> generalised disease, a finding supported by the work of Quilty et al.<br />

o In this latter study, 284 patients with <strong>prostate</strong> <strong>cancer</strong> and painful bone metastases were<br />

treated with local or hemi-body radiotherapy or strontium. Median survival was nonsignificantly<br />

different between groups (33 weeks with strontium versus 28 weeks with<br />

radiotherapy; p=0.1). 129<br />

o Both radiotherapy and strontium provided effective pain relief that was sustained <strong>for</strong><br />

3 months in 63.6% of patients after hemi-body radiotherapy compared with 66.1% of<br />

patients after strontium, and in 61% of patients after local radiotherapy compared with<br />

65.9% of patients in the comparable strontium group.<br />

o Fewer patients reported new pain sites after strontium than after local or hemi-body<br />

radiotherapy (p


Ongoing Support<br />

The <strong>MDT</strong> <strong>team</strong> should ensure regular communication with the primary care <strong>team</strong>.<br />

This may mean:<br />

• Timely provision of detailed discharge or outpatient summaries<br />

• Explanation of why a treatment route has been decided upon<br />

• The patient’s response to the chosen treatment<br />

• Sharing of protocols<br />

• Online educational resources<br />

• Agreement on prescribing policies<br />

• Provision of contact numbers <strong>for</strong> requests <strong>for</strong> in<strong>for</strong>mation<br />

The local patient support network, e.g. partner/family, must be included in the in<strong>for</strong>mation/education<br />

process through the use of:<br />

• Patient in<strong>for</strong>mation materials<br />

• Audio visual materials such as videos, DVDs and Web-based in<strong>for</strong>mation<br />

51


References<br />

1. Integrated Care Network. Integrated working: a guide. June, 2004. Available at: www.<br />

integratedcarenetwork.gov.uk/docs/publications/11/13.pdf.<br />

2. National Institute <strong>for</strong> Clinical Excellence. Guidance <strong>for</strong> commissioning <strong>cancer</strong> services. Improving<br />

outcomes in urological <strong>cancer</strong>s. The research evidence. London: NICE, 2002.<br />

3. House of Commons Committee of Public Accounts. The NHS Cancer Plan: a progress report.<br />

Twentieth report of session 2005−06. London: The Stationery Office, 2006.<br />

4. Davison BJ, Parker PA, Goldenberg SL. Patients’ preferences <strong>for</strong> communicating a <strong>prostate</strong> <strong>cancer</strong><br />

diagnosis and participating in medical decision-making. BJU Int 2004; 93: 47−51.<br />

5. Schroder FH, Hugosson Roobol MJ, et al. Screening and <strong>prostate</strong> <strong>cancer</strong> mortality in a randomized<br />

European study. N Engl J Med 2009; 360: 1320−1328.<br />

6. Andriole GL, Craw<strong>for</strong>d ED, Grubb RL 3rd, et al. Mortality results from a randomized <strong>prostate</strong><br />

<strong>cancer</strong> screening trial. N Engl J Med 2009; 360: 1310−1319.<br />

7. Rosario DJ, Lane JA, Metcalfe C, et al. Contribution of a single repeat PSA test to <strong>prostate</strong> cáncer<br />

risk assessment: experience from the ProtecT study. Eur Urol 2008; 53: 777−784.<br />

8. Ben-Shlomo Y, Evans S, Ibrahim F, et al. The risk of <strong>prostate</strong> <strong>cancer</strong> amongst black men in the<br />

United Kingdom: the PROCESS cohort study. Eur Urol 2008; 53: 99−105.<br />

9. Steinberg GD, Carter BS, Beaty TH, Childs B, Walsh PC. Family history and the risk of <strong>prostate</strong><br />

<strong>cancer</strong>. Prostate 1990; 17: 337−347.<br />

10. Negri E, Pelucchi C, Talamini R, et al. Family history of <strong>cancer</strong> and the risk of <strong>prostate</strong> <strong>cancer</strong> and<br />

benign prostatic hyperplasia. Int J Cancer 2005; 114: 648−652.<br />

11. Catalona WJ, Richie JP, DeKernion JB, et al. Comparison of <strong>prostate</strong> specific antigen concentration<br />

versus <strong>prostate</strong> specific antigen density in the early detection of <strong>prostate</strong> <strong>cancer</strong>: Receiver<br />

operating characteristic curves. J Urol 1994; 152 (6 Pt 1): 2031−2036.<br />

12. Elgamal A-AA, Cornillie FJ, Van Poppel HP, et al. Free-to-total <strong>prostate</strong> specific antigen ratio as a<br />

single test <strong>for</strong> detection of significant stage T1c <strong>prostate</strong> <strong>cancer</strong>. J Urol 1996; 156: 1042−1049.<br />

13. Aus G, Becker C, Lilja H, et al. Free-to-total <strong>prostate</strong>-specific antigen ratio as a predictor of nonorgan-confined<br />

<strong>prostate</strong> <strong>cancer</strong> (stage pT3). Scand J Urol Nephrol 2003; 37: 466−470.<br />

14. Aus G, Becker C, Franzen S, et al. Cumulative <strong>prostate</strong> <strong>cancer</strong> risk assessment with the aid of the<br />

free-to-total <strong>prostate</strong> specific antigen ratio. Eur Urol 2004; 45: 160−165.<br />

15. Hudson MA, Bahnson RR, Catalona WJ. Clinical use of <strong>prostate</strong> specific antigen in patients with<br />

<strong>prostate</strong> <strong>cancer</strong>. J Urol 1989; 142: 1011−1017.<br />

16. Brawer MK, Lange PH. Prostate-specific antigen in management of prostatic carcinoma. Urology<br />

1989; 33 (5 Suppl): 11−16.<br />

17. Partin AW, Carter HB, Chan DW, et al. Prostate specific antigen in the staging of localized <strong>prostate</strong><br />

<strong>cancer</strong>: influence of tumor differentiation, tumor volume and benign hyperplasia. J Urol 1990;<br />

143: 747−752.<br />

18. Dearnaley DP, Kirby RS, Kirk D, Malone P, Simpson RJ, Williams G. Diagnosis and management of<br />

early <strong>prostate</strong> <strong>cancer</strong>. Report of a British Association of Urological Surgeons working party. BJU Int<br />

1999; 83: 18−33.<br />

19. DeAntoni EP, Craw<strong>for</strong>d ED, Oesterling JE, et al. Age- and race-specific reference ranges <strong>for</strong><br />

<strong>prostate</strong>-specific antigen from a large community-based study. Urology 1996; 48: 234−239.<br />

20. Xu ZQ, Hua LX, Qian LX, Wu HF. Effect of transrectal prostatic biopsy on serum <strong>prostate</strong>-specific<br />

antigen levels. Zhonghua Nan Ke Xue 2002; 8: 341−342.<br />

52


21. Tchetgen MB, Oesterling JE. The effect of prostatitis, urinary retention, ejaculation and<br />

ambulation on the serum <strong>prostate</strong>-specific antigen concentration. Urol Clin North Am 1997; 24:<br />

283−291.<br />

22. Gamé X, Vincendeau S, Palascak R, Milcent S, Fournier R, Houlgatte A. Total and free <strong>prostate</strong>specific<br />

antigen levels during the first month of acute prostatitis. Eur Urol 2003; 43: 702−705.<br />

23. Catalona WJ, Partin AW, Slawin KM, et al. Use of the percentage of free <strong>prostate</strong>-specific antigen<br />

to enhance differentiation of <strong>prostate</strong> <strong>cancer</strong> from benign prostatic disease: A prospective<br />

<strong>multi</strong>center clinical trial. JAMA 1998; 279: 1542−1547.<br />

24. Smith DS, Catalona WJ. Rate of change of serum PSA levels as a method <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong><br />

detection: J Urol 1994; 152: 1163−1167.<br />

25. Gustafsson O, Norming U, Almgard LE, et al. Diagnostic methods in the detection of <strong>prostate</strong><br />

<strong>cancer</strong>: a study of a randomly selected population of 2,400 men. J Urol 1992; 148: 1827−1831.<br />

26. Mettlin C, Murphy GP, Babalan RJ, et al. The results of a five-year early <strong>prostate</strong> <strong>cancer</strong> detection<br />

intervention. Cancer 1996; 77: 150−159.<br />

27. Jones WT, Resnick MI. Prostate ultrasound in screening, diagnosis, and staging of <strong>prostate</strong> <strong>cancer</strong>.<br />

Prob Urol 1990; 4: 343−357.<br />

28. Ellis WJ, Chetner MP, Preston SD, Brawer MK. Diagnosis of prostatic carcinoma: The yield of serum<br />

<strong>prostate</strong> specific antigen, digital rectal examination and transrectal ultrasonography. J Urol 1994;<br />

152(5 I): 1520−1525.<br />

29. National Institute <strong>for</strong> Health and Clinical Excellence. Prostate <strong>cancer</strong>: diagnosis and treatment. Full<br />

guideline. London, NICE: 2008.<br />

30. Eskicorapci SY, Baydar DE, Akbal C, et al. An extended 10-core transrectal ultrasonography guided<br />

<strong>prostate</strong> biopsy protocol improves the detection of <strong>prostate</strong> <strong>cancer</strong>. Eur Urol 2004; 45: 444−448.<br />

31. Djavan B, Fong YK, Ravery V, et al. Are repeat biopsies required in men with PSA < or =4 ng/ml? A<br />

<strong>multi</strong>institutional prospective European study. Eur Urol 2005; 47: 38−44.<br />

32. Allen DJ, Hindley R, Clovis S, et al. Does body-coil magnetic-resonance imaging have a role in<br />

the preoperative staging of patients with clinically localized <strong>prostate</strong> <strong>cancer</strong>? BJU Int 2004; 94:<br />

534−538.<br />

33. Gerber G, Chodak GW. Assessment of value of routine bone scans in patients with newly<br />

diagnosed <strong>prostate</strong> <strong>cancer</strong>. Urology 1991; 37: 418−422.<br />

34. D’Amico AV, Whittington R, Malkowicz SB, et al. Biochemical outcome after radical <strong>prostate</strong>ctomy,<br />

external beam radiation therapy, or interstitial radiation therapy <strong>for</strong> clinically localized <strong>prostate</strong><br />

<strong>cancer</strong>. JAMA 1998; 280: 969−974.<br />

35. van As NJ, Norman AR, Thomas K, et al. Predicting the probability of deferred radical treatment<br />

<strong>for</strong> localised <strong>prostate</strong> <strong>cancer</strong> managed by active surveillance. Eur Urol 2008; 54: 1297−1305.<br />

36. van den Bergh RC, Roemeling S, Roobol MJ, et al. Outcomes of men with screen-detected <strong>prostate</strong><br />

<strong>cancer</strong> eligible <strong>for</strong> active surveillance who were managed expectantly. Eur Urol 2008; Sep 17<br />

[epub].<br />

37. Draisma G, Boer R, Otto SJ, et al. Lead times and overdetection due to <strong>prostate</strong>-specific antigen<br />

screening: estimates from the European Randomized Study of Screening <strong>for</strong> Prostate Cancer. J<br />

Natl Cancer Inst 2003; 95: 868−878.<br />

38. Selley S, Donovan J, Faulkner A, et al. Diagnosis, management and screening of early localised<br />

<strong>prostate</strong> <strong>cancer</strong>. Health Technol Assess 1997; 1: 1−96.<br />

53


39. Bill-Axelson A, Holmberg L, Ruutu M, et al. Radical <strong>prostate</strong>ctomy versus watchful waiting in early<br />

<strong>prostate</strong> <strong>cancer</strong>. N Engl J Med 2005; 352: 1977−1984.<br />

40. Singer PA, Tasch ES, Stocking C, et al. Sex or survival: trade-offs between quality and quantity of<br />

life. J Clin Oncol 1991; 9: 328−334.<br />

41. Bill-Axelson A, Holmberg L, Filen F, et al. Radical <strong>prostate</strong>ctomy versus watchful waiting in<br />

localized <strong>prostate</strong> <strong>cancer</strong>: the Scandinavian <strong>prostate</strong> <strong>cancer</strong> group-4 randomized trial. J Natl<br />

Cancer Inst 2008; 100: 1144−1154.<br />

42. Bonney WW, Schned AR, Timberlake DS. Neoadjuvant androgen ablation <strong>for</strong> localised prostatic<br />

<strong>cancer</strong>. Pathology methods, surgical end points and meta-analysis of randomised trials. J Urol<br />

1999; 160: 1754−1760.<br />

43. Paul R, Van Randenborgh H, Kubler H, et al. Significance of neoadjuvant therapy be<strong>for</strong>e radical<br />

<strong>prostate</strong>ctomy. Urologe A 2004; 43: 680−688.<br />

44. Selli C, Milesi C. Neoadjuvant androgen deprivation be<strong>for</strong>e radical <strong>prostate</strong>ctomy. A review.<br />

Minerva Urol Nefrol 2004; 56: 165−171.<br />

45. Witjes WPJ, Schulman CC, Debruyne FMJ. Preliminary results of a prospective randomised study<br />

comparing radical <strong>prostate</strong>ctomy versus radical <strong>prostate</strong>ctomy associated with neo-adjuvant<br />

hormonal combination therapy in T2-3 N0 M0 prostatic carcinoma. Urology 1997; 49 (3 Suppl):<br />

65−69.<br />

46. Hachiya T, Minei S, Hirano D, et al. Adjuvant hormone therapy in patients with positive surgical<br />

margins after radical <strong>prostate</strong>ctomy. Jpn J Urol 2002; 93: 469−475.<br />

47. Prayer-Galetti T, et al. Disease free survival in patients with pathological “C Stage” <strong>prostate</strong> <strong>cancer</strong><br />

at radical <strong>prostate</strong>ctomy submitted to adjuvant hormonal treatment. Eur Urol 2000; 38: 504, abs<br />

48.<br />

48. Bolla M, van Poppel H, Collette L, et al. Postoperative radiotherapy after radical <strong>prostate</strong>ctomy: a<br />

randomised controlled trial (EORTC trial 22911). Lancet 2005; 366: 572−578.<br />

49. Thompson Jr IM, Tangen CM, Paradelo J, et al. Adjuvant radiotherapy <strong>for</strong> pathologically advanced<br />

<strong>prostate</strong> <strong>cancer</strong>: a randomized clinical trial. JAMA 2006; 296: 2329−2335.<br />

50. Parker C, Sydes MR, Catton C, et al. Radiotherapy and androgen deprivation in combination after<br />

local surgery (RADICALS): A new Medical Research Council/National Cancer Institute of Canada<br />

phase III trial of adjuvant treatment after radical <strong>prostate</strong>ctomy. BJU Int 2007; 99: 1376−1379.<br />

51. Nilsson S, Norlen BJ, Widmark A. A systematic overview of radiation therapy effects in <strong>prostate</strong><br />

<strong>cancer</strong>. Acta Oncol 2004; 43: 316−381.<br />

52. Dearnaley DP, Khoo VS, Norman AR, et al. Comparison of radiation side-effects of con<strong>for</strong>mal and<br />

conventional radiotherapy in <strong>prostate</strong> <strong>cancer</strong>: a randomised trial. Lancet 1999; 353: 267−272.<br />

53. Pollack A, Zagars GK, Starkschall G, et al. Prostate <strong>cancer</strong> radiation dose response: results of the M.<br />

D. Anderson phase III randomized trial. Int J Radiat Oncol Biol Phys 2002; 53: 1097−1105.<br />

54. Dearnaley DP, Sydes MR, Graham JD, et al. Escalated-dose versus standard-dose con<strong>for</strong>mal<br />

radiotherapy in <strong>prostate</strong> <strong>cancer</strong>: first results from the MRC RT01 randomised controlled trial.<br />

Lancet Oncol 2007; 8: 475−487.<br />

55. Zelefsky MJ, Fuks Z, Lee HJ, et al. High dose radiation delivered by intensity modulated con<strong>for</strong>mal<br />

radiotherapy improves the outcome of localized <strong>prostate</strong> <strong>cancer</strong>. J Urol 2001; 166: 876−878.<br />

56. Zelefsky MJ, Fuks Z, Hunt M, et al. High-dose intensity modulated radiation therapy <strong>for</strong> <strong>prostate</strong><br />

<strong>cancer</strong>: early toxicity and biochemical outcome in 772 patients. Int J Radiat Oncol Biol Phys 2002;<br />

52: 1111−1116.<br />

54


57. Amer AM, Mott J, Mackay RI, et al. Prediction of the benefits from dose-escalated<br />

hypofractionated intensity-modulated radiotherapy <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong>. Int J Radiat Oncol Biol<br />

Phys 2003; 56: 199−207.<br />

58. South CP, Khoo VS, Naismith O, Norman A, Dearnaley DP. A comparison of treatment planning<br />

techniques used in two randomised UK external beam radiotherapy trials <strong>for</strong> localised <strong>prostate</strong><br />

<strong>cancer</strong>. Clin Oncol (R Coll Radiol) 2008; 20: 15−21.<br />

59. Shearer RJ, Davies JH, Gelister JS, Dearnaley DP. Hormonal cytoreduction and radiotherapy <strong>for</strong><br />

carcinoma of the <strong>prostate</strong>. Br J Urol 1992; 69: 521−524.<br />

60. Forman JD, Kumar R, Haas G, et al. Neoadjuvant hormonal downsizing of localized carcinoma of<br />

the <strong>prostate</strong>: effects on the volume of normal tissue irradiation. Cancer Invest 1995; 13: 8−15.<br />

61. Hara I, Miyake H, Yamada Y. Neoadjuvant androgen withdrawal prior to external radiotherapy <strong>for</strong><br />

locally advanced adenocarcinoma of the <strong>prostate</strong>. Int J Urol 2002; 9: 322−332.<br />

62. Pilepich MV, Winter K, John MJ, et al. Phase III RTOG trial 86-10 of androgen deprivation adjuvant<br />

to definitive radiotherapy in locally advanced carcinoma of the <strong>prostate</strong>. Int J Radiat Oncol Biol<br />

Phys 2001; 50: 1243−1252.<br />

63. Roach M 3rd, Bae K, Speight J, et al. Short-term neoadjuvant androgen deprivation therapy and<br />

external-beam radiotherapy <strong>for</strong> locally advanced <strong>prostate</strong> <strong>cancer</strong>: long-term results of RTOG 8610.<br />

J Clin Oncol 2008; 26: 585−591.<br />

64. Denham JW, Steigler A, Wilcox C, et al. Time to biochemical failure and <strong>prostate</strong>-specific antigen<br />

doubling time as surrogates <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong>-specific mortality: evidence from the TROG 96.01<br />

randomised controlled trial. Lancet Oncol 2008; 9: 1058−1068.<br />

65. D’Amico AV, Manola J, Loffredo M, Renshaw AA, DellaCroce A, Kantoff PW. 6-month androgen<br />

suppression plus radiation therapy vs radiation therapy alone <strong>for</strong> patients with clinically localized<br />

<strong>prostate</strong> <strong>cancer</strong>: a randomized controlled trial. JAMA 2004; 292: 821−827.<br />

66. National Institute <strong>for</strong> Health and Clinical Excellence. Low dose rate brachytherapy <strong>for</strong> localised<br />

<strong>prostate</strong> <strong>cancer</strong>. Interventional Procedure Guidance 132, July 2005.<br />

67. Crook J, Lukka H, Klotz L, et al. Systematic overview of the evidence <strong>for</strong> brachytherapy in clinically<br />

localised <strong>prostate</strong> <strong>cancer</strong>. CMAJ 2001; 164: 975−981.<br />

68. Merrick GS, Butler WM, Lief JH, Dorsey AT. Is brachytherapy comparable with radical<br />

<strong>prostate</strong>ctomy and external beam radiotherapy <strong>for</strong> clinically localised <strong>prostate</strong> <strong>cancer</strong>? Tech Urol<br />

2001; 7: 12−19.<br />

69. Singh AM, Gagnon G, Collins B, et al. Combined external beam radiotherapy and Pd-103<br />

brachytherapy boost improves biochemical failure free survival in patients with clinically localized<br />

<strong>prostate</strong> <strong>cancer</strong>: results of a matched pair analysis. Prostate 2005; 62: 54−60.<br />

70. Potters L, Morgenstern C, Calugaru E, et al. 12-year outcomes following permanent <strong>prostate</strong><br />

brachytherapy in patients with clinically localized <strong>prostate</strong> <strong>cancer</strong>. J Urol 2005; 173: 1562−1566.<br />

71. Wills F, Hailey D. Brachytherapy <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong>. Alberta Heritage Foundation <strong>for</strong> Medical<br />

Research, Alberta Canada. 1999, Dec. (http://www.ahfmr.ab.ca/publications).<br />

72. Beyer DC, Priestley JB Jr. Biochemical disease-free survival following 125I <strong>prostate</strong> implantation.<br />

Int J Radiat Oncol Biol Phys 1997; 37: 559−563.<br />

73. Blasko JC, Wallner K, Grimm PD, Ragde H. Prostate specific antigen based disease control<br />

following ultrasound guided 125iodine implantation <strong>for</strong> stage T1/T2 prostatic carcinoma. J Urol<br />

1995; 154: 1096−1099.<br />

74. Wallner K, Roy J, Zelefsky M, Harrison L. Short-term freedom from disease progression after I-125<br />

<strong>prostate</strong> implantation. Int J Radiat Oncol Biol Phys 1994; 30: 405−409.<br />

55


75. Stock RG, Stone NN, DeWyngaert JK, Lavagnini P, Unger PD. Prostate specific antigen findings and<br />

biopsy results following interactive ultrasound guided transperineal brachytherapy <strong>for</strong> early stage<br />

<strong>prostate</strong> carcinoma. Cancer 1996; 77: 2386−2392.<br />

76. Beyer DC, Brachman DG. Failure free survival following brachytherapy alone <strong>for</strong> <strong>prostate</strong> <strong>cancer</strong>:<br />

comparison with external beam radiotherapy. Radiother Oncol 2000; 57: 263−267.<br />

77. Ragde H, Korb LJ, Elgamal A-A, et al. Modern <strong>prostate</strong> brachytherapy. PSA results in 219 patients<br />

with up to 12 years of observational follow up. Cancer 2000; 89: 135−141.<br />

78. Kupelian PA, Potters L, Khuntia D, et al. Radical <strong>prostate</strong>ctomy, external beam radiotherapy or =72 Gy, permanent seed implantation, or combined seeds/<br />

external beam radiotherapy <strong>for</strong> stage T1-T2 <strong>prostate</strong> <strong>cancer</strong>. Int J Radiat Oncol Biol Phys 2004; 58:<br />

25−33.<br />

79. Han KR, Cohen JK, Miller RJ, et al. Treatment of organ confined <strong>prostate</strong> <strong>cancer</strong> with third<br />

generation cryosurgery: preliminary <strong>multi</strong>center experience. J Urol 2003; 170 (4 Pt 1): 1126−1130.<br />

80. Zisman A, Pantuck AJ, Cohen JK, Belldegrun AS. Prostate cryoablation using direct transperineal<br />

placement of ultrathin probes through a 17-gauge brachytherapy template-technique and<br />

preliminary results. Urology 2001; 58: 988−993.<br />

81. Parker MC, Cook A, Riddle PR, et al. Is delayed treatment justified in carcinoma of the <strong>prostate</strong>? Br<br />

J Urol 1985; 57: 724−728.<br />

82. Rana A, Chisholm GD, Khan M, et al. Conservative management with symptomatic treatment<br />

and delayed hormonal manipulation is justified in men with locally advanced carcinoma of the<br />

<strong>prostate</strong>. Br J Urol 1994; 74: 637−641.<br />

83. Wilt T, Nair B, MacDonald R, Rutks I. Early versus deferred androgen suppression in the treatment<br />

of advanced prostatic <strong>cancer</strong>. Cochrane Database Syst Rev 2001; CD003506.<br />

84. Adolfsson J, Steineck G, Hedlund PO. Deferred treatment of locally advanced nonmetastatic<br />

<strong>prostate</strong> <strong>cancer</strong>: a long-term followup. J Urol 1999; 161: 505−508.<br />

85. MRC Prostate Working Party Investigators Group. Immediate versus deferred treatment <strong>for</strong><br />

advanced <strong>prostate</strong> <strong>cancer</strong>. Initial results of the MRC trial. Br J Urol 1997; 79: 235−246.<br />

86. Studer UE, Whelan P, Albrecht W, et al. Immediate or deferred androgen deprivation <strong>for</strong> patients<br />

with <strong>prostate</strong> <strong>cancer</strong> not suitable <strong>for</strong> local treatment with curative intent: European Organisation<br />

<strong>for</strong> Research and Treatment of Cancer (EORTC) Trial 30891. J Clin Oncol 2006; 24: 1868−1876.<br />

87. McLeod DG, Iversen P, See WA, et al. Bicalutamide 150 mg plus standard care versus standard care<br />

alone <strong>for</strong> early <strong>prostate</strong> <strong>cancer</strong>. BJU Int 2006; 97: 247−254.<br />

88. Widmark A, Klepp O, Solberg A, et al. Endocrine treatment, with or without radiotherapy, in<br />

locally advanced <strong>prostate</strong> <strong>cancer</strong> (SPCG-7/SFUO-3): an open randomised phase III trial. Lancet 2009;<br />

373: 301−308.<br />

89. Lawton CA, DeSilvio M, Roach M 3rd, et al. An update of the phase III trial comparing whole pelvic<br />

to <strong>prostate</strong> only radiotherapy and neoadjuvant to adjuvant total androgen suppression: updated<br />

analysis of RTOG 94-13, with emphasis on unexpected hormone/radiation interactions. Int J Radiat<br />

Oncol Biol Phys 2007; 69: 646−655.<br />

90. Martinez AA, Gustafson G, Gonzalez J, et al. Dose escalation using con<strong>for</strong>mal high dose rate<br />

brachytherapy improves outcome in unfavorable <strong>prostate</strong> <strong>cancer</strong>. Int J Radiat Oncol Biol Phys<br />

2002; 53: 316−327.<br />

91. Bolla M, Collette L, Blank L, et al. Long-term results with immediate androgen suppression and<br />

external irradiation in patients with locally advanced <strong>prostate</strong> <strong>cancer</strong> (an EORTC study): a phase III<br />

randomised trial. Lancet 2002; 360: 103−106.<br />

56


92. Bolla M, Collette L, Van Tienhoven G, et al. Ten year results of long term adjuvant androgen<br />

deprivation with goserelin in patients with locally advanced <strong>prostate</strong> <strong>cancer</strong> treated with<br />

radiotherapy: A phase III EORTC study. Int J Radiat Oncol Biol Phys 2008; 72 (1 Suppl1): S30−S31.<br />

93. Bolla M, de Reijke TM, Van Tienhoven G, et al. Duration of androgen suppression in the treatment<br />

of <strong>prostate</strong> <strong>cancer</strong>. N Engl J Med 2009; 360: 2516−2527.<br />

94. Pilepich MV, Winter K, Lawton CA, et al. Androgen suppression adjuvant to definitive<br />

radiotherapy in <strong>prostate</strong> carcinoma − long-term results of phase III RTOG 85-31. Int J Radiat Oncol<br />

Biol Phys 2005; 61: 1285−1290.<br />

95. Hanks GE, Pajak TF, Porter A, et al. Phase III trial of long-term adjuvant androgen deprivation<br />

after neoadjuvant hormonal cytoreduction and radiotherapy in locally advanced carcinoma of the<br />

<strong>prostate</strong>: the Radiation Therapy Oncology Group Protocol 92-02. J Clin Oncol 2003; 21: 3972−3978.<br />

96. Horwitz EM, Bae K, Hanks GE, et al. Ten-year follow-up of radiation therapy oncology group<br />

protocol 92-02: a phase III trial of the duration of elective androgen deprivation in locally<br />

advanced <strong>prostate</strong> <strong>cancer</strong>. J Clin Oncol 2008; 26: 2497−2504.<br />

97. Roach M, Hanks G, Thames H, et al. Defining biochemical failure following radiotherapy with or<br />

without hormonal therapy in men with clinically localized <strong>prostate</strong> <strong>cancer</strong>: recommendations of<br />

the RTOG-ASTRO Phoenix Consensus Conference. Int J Radiat Oncol Biol Phys 2006; 65: 965−974.<br />

98. Pound CR, Partin AW, Eisenberger MA, et al. Natural history of progression after PSA elevation<br />

following radical <strong>prostate</strong>ctomy. JAMA 1999; 281: 1591−1597.<br />

99. Messing EM, Manola J, Yao J, et al. Immediate versus deferred androgen deprivation<br />

treatment in patients with node-positive <strong>prostate</strong> <strong>cancer</strong> after radical <strong>prostate</strong>ctomy and pelvic<br />

lymphadenectomy. Lancet Oncol 2006; 7: 472−479.<br />

100. Touma NJ, Izawa JI, Chin JL. Current status of local salvage therapies following radiation failure<br />

<strong>for</strong> <strong>prostate</strong> <strong>cancer</strong>. J Urol 2005; 173: 373−379.<br />

101. Grado GL, Collins JM, Kriegshauser, JS et al. Salvage brachytherapy <strong>for</strong> localized <strong>prostate</strong> <strong>cancer</strong><br />

after radiotherapy failure. Urology 1999; 53: 2−10.<br />

102. Gelet A, Chapelon JY, Poissonier L, et al. Local recurrence of <strong>prostate</strong> <strong>cancer</strong> after external beam<br />

radiotherapy: early experience of salvage therapy using high-intensity focused ultrasonography.<br />

Urology 2004; 63: 625−629.<br />

103. Tombal B. The importance of testosterone control in <strong>prostate</strong> <strong>cancer</strong>. Eur Urol Suppl 2007; 6:<br />

834−839.<br />

104. Vogelzang NJ, Chodak GW, Soloway MS, et al. Goserelin versus orchiectomy in the treatment<br />

of advanced <strong>prostate</strong> <strong>cancer</strong>: final results of a randomized trial. Zoladex Prostate Study Group.<br />

Urology 1995; 46: 220−226.<br />

105. Kaisary AV, Tyrrell CJ, Peeling WB, Griffiths K. Comparison of LHRH analogue (Zoladex) with<br />

orchiectomy in patients with metastatic prostatic carcinoma. Br J Urol 1991; 67: 502−508.<br />

106. Seidenfeld J, Samson DJ, Hasselblad V, et al. Single-therapy androgen suppression in men with<br />

advanced <strong>prostate</strong> <strong>cancer</strong>: a systematic review and meta-analysis. Ann Intern Med 2000; 132:<br />

566−577.<br />

107. Shahinian VB, Kuo YF, Freeman JL, Orihuela E, Goodwin JS. Increasing use of gonadotropinreleasing<br />

hormone agonists <strong>for</strong> the treatment of localized <strong>prostate</strong> carcinoma. Cancer 2005; 103:<br />

1615−2164.<br />

108. Kaku H, Saika T, Tsushima T, et al. Time course of serum testosterone and luteinizing hormone<br />

levels after cessation of long-term luteinizing hormone-releasing hormone agonist treatment in<br />

patients with <strong>prostate</strong> <strong>cancer</strong>. Prostate 2006; 66: 439−444.<br />

57


109. Cassileth BR, Soloway MS, Vogelzang NJ, et al. Quality of life and psychosocial status in stage D<br />

<strong>prostate</strong> <strong>cancer</strong>. Zoladex Prostate Cancer Study Group. Qual Life Res 1992; 1: 323−330.<br />

110. Klotz L, Boccon-Gibod L, Shore ND, et al. The efficacy and safety of degarelix: a 12-month<br />

comparative, randomized, open-label, parallel-group phase III study in patients with <strong>prostate</strong><br />

<strong>cancer</strong>. BJU Int 2008; 102: 1531−1538.<br />

111. Prostate Cancer Trialists’ Collaborative Group. Maximum androgen blockade in advanced <strong>prostate</strong><br />

<strong>cancer</strong>: an overview of the randomised trials. Lancet 2000; 355: 1491−1498.<br />

112. Klotz L. Combined androgen blockade in <strong>prostate</strong> <strong>cancer</strong>: Meta-analyses and associated issues.<br />

BJU Int 2001; 87: 806−813.<br />

113. Sartor AO, Tangen CM, Hussain MH, et al. Antiandrogen withdrawal in castrate-refractory<br />

<strong>prostate</strong> <strong>cancer</strong>: a Southwest Oncology Group trial (SWOG 9426). Cancer 2008; 112: 2393−2400.<br />

114. Byar DP. Treatment of prostatic <strong>cancer</strong>: studies by the Veterans Administration Cooperative<br />

Urological Research Group. Bull N Y Acad Med 1972; 48: 751−766.<br />

115. Robinson MR, Smith PH, Richards B, et al. The final analysis of the EORTC Genito-Urinary Tract<br />

Cancer Co-Operative Group phase III clinical trial (protocol 30805) comparing orchidectomy,<br />

orchidectomy plus cyproterone acetate and low dose stilboestrol in the management of<br />

metastatic carcinoma of the <strong>prostate</strong>. Eur Urol 1995; 28: 273−283.<br />

116. Tannock IF, Osoba D, Stockler MR, et al. Chemotherapy with mitoxantrone plus prednisone or<br />

prednisone alone <strong>for</strong> symptomatic hormone-resistant <strong>prostate</strong> <strong>cancer</strong>: a Canadian randomized<br />

trial with palliative end points. J Clin Oncol 1996; 14: 1756−1764.<br />

117. Small EJ, Halabi S, Dawson NA, et al. Antiandrogen withdrawal alone or in combination with<br />

ketoconazole in androgen-independent <strong>prostate</strong> <strong>cancer</strong> patients: a phase III trial (CALGB 9583).<br />

J Clin Oncol 2004; 22: 1025−1033.<br />

118. Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone<br />

<strong>for</strong> advanced <strong>prostate</strong> <strong>cancer</strong>. N Engl J Med 2004; 351: 1502−1512.<br />

119. Berthold DR, Pond GR, Soban F, de Wit R, Eisenberger M, Tannock I. Docetaxel plus prednisone<br />

or mitoxantrone plus prednisone <strong>for</strong> advanced <strong>prostate</strong> <strong>cancer</strong>: updated survival in the TAX 327<br />

study. J Clin Oncol 2008; 26: 242−245.<br />

120. Porter AT, McEwan AJ, Powe JE, et al. Results of a randomised phase III trial to evaluate the<br />

efficacy of strontium-89 adjuvant to local field EB-RT in the management of endocrine resistant<br />

metastatic <strong>prostate</strong> <strong>cancer</strong>. Int J Radiat Oncol Biol Phys 1993; 25: 805−813.<br />

121. Brundage MD, Crook JM, Lukka H, et al. Use of strontium-89 in endocrine refractory <strong>prostate</strong><br />

<strong>cancer</strong> metastatic to bone. Provincial genitourinary <strong>cancer</strong> disease group. Cancer Prev Control<br />

1998; 2: 79−87.<br />

122. Robinson RG, Preston DF, Schiefelbein M, Baxter KG. Strontium–89 therapy <strong>for</strong> the palliation of<br />

pain due to osseous metastases. JAMA 1995; 274: 420−424.<br />

123. Tu SM, Millikan RE, Mengistu B, et al. bone targeted therapy <strong>for</strong> advanced and androgen<br />

independent <strong>prostate</strong> <strong>cancer</strong>. A randomised phase II trial. Lancet 2001; 357: 336−341.<br />

124. Saad F, Gleason DM, Murray R, et al. A randomized, placebo-controlled trial of zoledronic acid<br />

in patients with hormone-refractory metastatic <strong>prostate</strong> carcinoma. J Natl Cancer Inst 2002; 94:<br />

1458−1468.<br />

125. Dearnaley DP, Mason MD, Parmar MK, Sanders K, Sydes MR. Adjuvant therapy with oral sodium<br />

clodronate in locally advanced and metastatic <strong>prostate</strong> <strong>cancer</strong>: long-term overall survival results<br />

from the MRC PR04 and PR05 randomised controlled trials. Lancet Oncol 2009; 10: 872−876.<br />

58


126. Marx RE, Sawatari Y, Fortin M, Broumand V. Bisphosphonate-induced exposed bone<br />

(osteonecrosis/osteopetrosis) of the jaws: risk factors, recognition, prevention and treatment.<br />

J Oral Maxillofac Surg 2005; 63: 1567−1575.<br />

127. Zometa (zoledronic acid). Summary of product characteristics, July 2008.<br />

128. McQuay HJ, Carroll D, Moore RA. Radiotherapy <strong>for</strong> painful bone metastases. A systematic review.<br />

Clin Oncol 1997; 9: 150−154.<br />

129. Quilty PM, Kirk D, Bolger JJ, et al. A comparison of the palliative effects of strontium-89 and<br />

external beam radiotherapy in metastatic <strong>prostate</strong> <strong>cancer</strong>. Radiother Oncol 1994; 31: 33−40.<br />

59

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!